Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
bioRxiv ; 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38585848

RESUMO

RNA binding proteins (RBPs) containing intrinsically disordered regions (IDRs) are present in diverse molecular complexes where they function as dynamic regulators. Their characteristics promote liquid-liquid phase separation (LLPS) and the formation of membraneless organelles such as stress granules and nucleoli. IDR-RBPs are particularly relevant in the nervous system and their dysfunction is associated with neurodegenerative diseases and brain tumor development. SERBP1 is a unique member of this group, being mostly disordered and lacking canonical RNA-binding domains. Using a proteomics approach followed by functional analysis, we defined SERBP1's interactome. We uncovered novel SERBP1 roles in splicing, cell division, and ribosomal biogenesis and showed its participation in pathological stress granules and Tau aggregates in Alzheimer's disease brains. SERBP1 preferentially interacts with other G-quadruplex (G4) binders, implicated in different stages of gene expression, suggesting that G4 binding is a critical component of SERBP1 function in different settings. Similarly, we identified important associations between SERBP1 and PARP1/polyADP-ribosylation (PARylation). SERBP1 interacts with PARP1 and its associated factors and influences PARylation. Moreover, protein complexes in which SERBP1 participates contain mostly PARylated proteins and PAR binders. Based on these results, we propose a feedback regulatory model in which SERBP1 influences PARP1 function and PARylation, while PARylation modulates SERBP1 functions and participation in regulatory complexes.

2.
RNA Biol ; 20(1): 311-322, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-37294214

RESUMO

The exon junction complex (EJC) plays key roles throughout the lifespan of RNA and is particularly relevant in the nervous system. We investigated the roles of two EJC members, the paralogs MAGOH and MAGOHB, with respect to brain tumour development. High MAGOH/MAGOHB expression was observed in 14 tumour types; glioblastoma (GBM) showed the greatest difference compared to normal tissue. Increased MAGOH/MAGOHB expression was associated with poor prognosis in glioma patients, while knockdown of MAGOH/MAGOHB affected different cancer phenotypes. Reduced MAGOH/MAGOHB expression in GBM cells caused alterations in the splicing profile, including re-splicing and skipping of multiple exons. The binding profiles of EJC proteins indicated that exons affected by MAGOH/MAGOHB knockdown accumulated fewer complexes on average, providing a possible explanation for their sensitivity to MAGOH/MAGOHB knockdown. Transcripts (genes) showing alterations in the splicing profile are mainly implicated in cell division, cell cycle, splicing, and translation. We propose that high MAGOH/MAGOHB levels are required to safeguard the splicing of genes in high demand in scenarios requiring increased cell proliferation (brain development and GBM growth), ensuring efficient cell division, cell cycle regulation, and gene expression (splicing and translation). Since differentiated neuronal cells do not require increased MAGOH/MAGOHB expression, targeting these paralogs is a potential option for treating GBM.


Assuntos
Genes cdc , Glioblastoma , Humanos , Splicing de RNA , Divisão Celular , Núcleo Celular/metabolismo , Glioblastoma/metabolismo , Proteínas Nucleares/metabolismo
3.
Neuro Oncol ; 25(3): 459-470, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35862252

RESUMO

BACKGROUND: The loss of neurogenic tumor suppressor microRNAs miR-124, miR-128, and miR-137 is associated with glioblastoma's undifferentiated state. Most of their impact comes via the repression of a network of oncogenic transcription factors. We conducted a high-throughput functional siRNA screen in glioblastoma cells and identify E74 like ETS transcription factor 4 (ELF4) as the leading contributor to oncogenic phenotypes. METHODS: In vitro and in vivo assays were used to assess ELF4 impact on cancer phenotypes. We characterized ELF4's mechanism of action via genomic and lipidomic analyses. A MAPK reporter assay verified ELF4's impact on MAPK signaling, and qRT-PCR and western blotting were used to corroborate ELF4 regulatory role on most relevant target genes. RESULTS: ELF4 knockdown resulted in significant proliferation delay and apoptosis in GBM cells and long-term growth delay and morphological changes in glioma stem cells (GSCs). Transcriptomic analyses revealed that ELF4 controls two interlinked pathways: 1) Receptor tyrosine kinase signaling and 2) Lipid dynamics. ELF4 modulation directly affected receptor tyrosine kinase (RTK) signaling, as mitogen-activated protein kinase (MAPK) activity was dependent upon ELF4 levels. Furthermore, shotgun lipidomics revealed that ELF4 depletion disrupted several phospholipid classes, highlighting ELF4's importance in lipid homeostasis. CONCLUSIONS: We found that ELF4 is critical for the GBM cell identity by controlling genes of two dependent pathways: RTK signaling (SRC, PTK2B, and TNK2) and lipid dynamics (LRP1, APOE, ABCA7, PLA2G6, and PITPNM2). Our data suggest that targeting these two pathways simultaneously may be therapeutically beneficial to GBM patients.


Assuntos
Neoplasias Encefálicas , Glioblastoma , MicroRNAs , Humanos , Fatores de Transcrição/genética , Glioblastoma/patologia , MicroRNAs/genética , Receptores Proteína Tirosina Quinases/genética , Regulação Neoplásica da Expressão Gênica , Lipídeos , Proliferação de Células , Linhagem Celular Tumoral , Neoplasias Encefálicas/patologia , Proteínas de Ligação a DNA/genética , Proteínas Tirosina Quinases/metabolismo
4.
Cell Rep ; 36(1): 109254, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34233189

RESUMO

BAF chromatin remodeling complexes play important roles in chromatin regulation and cancer. Here, we report that Ewing sarcoma cells are dependent on the autocrine signaling mediated by NELL2, a secreted glycoprotein that has been characterized as an axon guidance molecule. NELL2 uses Robo3 as the receptor to transmit critical growth signaling. NELL2 signaling inhibits cdc42 and upregulates BAF complexes and EWS-FLI1 transcriptional output. We demonstrate that cdc42 is a negative regulator of BAF complexes, inducing actin polymerization and complex disassembly. Furthermore, we identify NELL2highCD133highEWS-FLI1high and NELL2lowCD133lowEWS-FLI1low populations in Ewing sarcoma, which display phenotypes consistent with high and low NELL2 signaling, respectively. We show that NELL2, CD133, and EWS-FLI1 positively regulate each other and upregulate BAF complexes and cell proliferation in Ewing sarcoma. These results reveal a signaling pathway regulating critical chromatin remodeling complexes and cancer cell proliferation.


Assuntos
Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/metabolismo , Antígeno AC133/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Montagem e Desmontagem da Cromatina , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos SCID , Proteínas de Fusão Oncogênica/metabolismo , Fenótipo , Polimerização , Subunidades Proteicas/metabolismo , Proteômica , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Receptores de Superfície Celular/metabolismo , Sarcoma de Ewing/genética , Regulação para Cima
5.
Cancers (Basel) ; 13(7)2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33804958

RESUMO

RNA-binding proteins (RBPs) function as master regulators of gene expression. Alterations in their levels are often observed in tumors with numerous oncogenic RBPs identified in recent years. Musashi1 (Msi1) is an RBP and stem cell gene that controls the balance between self-renewal and differentiation. High Msi1 levels have been observed in multiple tumors including glioblastoma and are often associated with poor patient outcomes and tumor growth. A comprehensive genomic analysis identified a network of cell cycle/division and DNA replication genes and established these processes as Msi1's core regulatory functions in glioblastoma. Msi1 controls this gene network via two mechanisms: direct interaction and indirect regulation mediated by the transcription factors E2F2 and E2F8. Moreover, glioblastoma lines with Msi1 knockout (KO) displayed increased sensitivity to cell cycle and DNA replication inhibitors. Our results suggest that a drug combination strategy (Msi1 + cell cycle/DNA replication inhibitors) could be a viable route to treat glioblastoma.

6.
Cells ; 11(1)2021 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-35011618

RESUMO

Medulloblastoma is the most common malignant brain tumor in children. Treatment with surgery, irradiation, and chemotherapy has improved survival in recent years, but patients are frequently left with devastating neurocognitive and other sequelae. Patients in molecular subgroups 3 and 4 still experience a high mortality rate. To identify new pathways contributing to medulloblastoma development and create new routes for therapy, we have been studying oncogenic RNA-binding proteins. We defined Musashi1 (Msi1) as one of the main drivers of medulloblastoma development. The high expression of Msi1 is prevalent in Group 4 and correlates with poor prognosis while its knockdown disrupted cancer-relevant phenotypes. Genomic analyses (RNA-seq and RIP-seq) indicated that cell cycle and division are the main biological categories regulated by Msi1 in Group 4 medulloblastoma. The most prominent Msi1 targets include CDK2, CDK6, CCND1, CDKN2A, and CCNA1. The inhibition of Msi1 with luteolin affected the growth of CHLA-01 and CHLA-01R Group 4 medulloblastoma cells and a synergistic effect was observed when luteolin and the mitosis inhibitor, vincristine, were combined. These findings indicate that a combined therapeutic strategy (Msi1 + cell cycle/division inhibitors) could work as an alternative to treat Group 4 medulloblastoma.


Assuntos
Ciclo Celular/genética , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Regulação Neoplásica da Expressão Gênica , Meduloblastoma/genética , Meduloblastoma/patologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Luteolina/farmacologia , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Fenótipo , Prognóstico , Vincristina/farmacologia , Quinases da Família src/metabolismo
7.
Sci Rep ; 10(1): 13399, 2020 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-32753612

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Genome Biol ; 21(1): 195, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32762776

RESUMO

BACKGROUND: RNA-binding proteins (RBPs) function as master regulators of gene expression. Alterations in RBP expression and function are often observed in cancer and influence critical pathways implicated in tumor initiation and growth. Identification and characterization of oncogenic RBPs and their regulatory networks provide new opportunities for targeted therapy. RESULTS: We identify the RNA-binding protein SERBP1 as a novel regulator of glioblastoma (GBM) development. High SERBP1 expression is prevalent in GBMs and correlates with poor patient survival and poor response to chemo- and radiotherapy. SERBP1 knockdown causes delay in tumor growth and impacts cancer-relevant phenotypes in GBM and glioma stem cell lines. RNAcompete identifies a GC-rich region as SERBP1-binding motif; subsequent genomic and functional analyses establish SERBP1 regulation role in metabolic routes preferentially used by cancer cells. An important consequence of these functions is SERBP1 impact on methionine production. SERBP1 knockdown decreases methionine levels causing a subsequent reduction in histone methylation as shown for H3K27me3 and upregulation of genes associated with neurogenesis, neuronal differentiation, and function. Further analysis demonstrates that several of these genes are downregulated in GBM, potentially through epigenetic silencing as indicated by the presence of H3K27me3 sites. CONCLUSIONS: SERBP1 is the first example of an RNA-binding protein functioning as a central regulator of cancer metabolism and indirect modulator of epigenetic regulation in GBM. By bridging these two processes, SERBP1 enhances glioma stem cell phenotypes and contributes to GBM poorly differentiated state.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Neoplasias Encefálicas/etiologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Epigênese Genética , Feminino , Glioblastoma/etiologia , Glioblastoma/mortalidade , Glioblastoma/terapia , Humanos , Masculino , Camundongos , Neurogênese , Fenótipo , Prognóstico , Estados Unidos/epidemiologia
9.
Sci Rep ; 10(1): 8979, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32488114

RESUMO

High-dose radiation is the main component of glioblastoma therapy. Unfortunately, radio-resistance is a common problem and a major contributor to tumor relapse. Understanding the molecular mechanisms driving response to radiation is critical for identifying regulatory routes that could be targeted to improve treatment response. We conducted an integrated analysis in the U251 and U343 glioblastoma cell lines to map early alterations in the expression of genes at three levels: transcription, splicing, and translation in response to ionizing radiation. Changes at the transcriptional level were the most prevalent response. Downregulated genes are strongly associated with cell cycle and DNA replication and linked to a coordinated module of expression. Alterations in this group are likely driven by decreased expression of the transcription factor FOXM1 and members of the E2F family. Genes involved in RNA regulatory mechanisms were affected at the mRNA, splicing, and translation levels, highlighting their importance in radiation-response. We identified a number of oncogenic factors, with an increased expression upon radiation exposure, including BCL6, RRM2B, IDO1, FTH1, APIP, and LRIG2 and lncRNAs NEAT1 and FTX. Several of these targets have been previously implicated in radio-resistance. Therefore, antagonizing their effects post-radiation could increase therapeutic efficacy. Our integrated analysis provides a comprehensive view of early response to radiation in glioblastoma. We identify new biological processes involved in altered expression of various oncogenic factors and suggest new target options to increase radiation sensitivity and prevent relapse.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma/genética , Glioblastoma/radioterapia , Splicing de RNA/genética , Transcrição Gênica/genética , Neoplasias Encefálicas/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Replicação do DNA/genética , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Expressão Gênica , Glioblastoma/patologia , Humanos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Biossíntese de Proteínas/genética , Tolerância a Radiação , Radiação Ionizante , Dosagem Radioterapêutica
10.
Physiol Genomics ; 48(5): 345-60, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26884460

RESUMO

Dynamic, epigenetic mechanisms can regulate macrophage phenotypes following exposure to different stimulating conditions and environments. However, temporal patterns of microRNAs (miRNAs or miRs) across multiple macrophage polarization phenotypes have not been defined. We determined miRNA expression in bone marrow-derived murine macrophages over multiple time points (0.5, 1, 3, 24 h) following exposure to cytokines and/or LPS. We hypothesized that dynamic changes in miRNAs regulate macrophage phenotypes. Changes in macrophage polarization markers were detected as early as 0.5 and as late as 24 h; however, robust responses for most markers occurred within 3 h. In parallel, many polarization-specific miRNAs were also changed by 3 h and expressed divergent patterns between M1 and M2a conditions, with increased expression in M1 (miR-155, 199a-3p, 214-3p, 455-3p, and 125a) or M2a (miR-511 and 449a). Specifically, miR-125a-5p exhibited divergent patterns: increased at 12-24 h in M1 macrophages and decreasing trend in M2a. VEGF in the culture media of macrophages was dependent upon the polarization state, with greatly diminished VEGF in M2a compared with M1 macrophage culture media despite similar VEGF in cell lysates. Inhibition of miR-125a-5p in media-only controls (MO) and M1 macrophages greatly increased expression and secretion of soluble VEGF receptor-1 (sVEGFR1) leading to diminished VEGF in the culture media, partially converting MO and M1 into an M2a phenotype. Thus, the divergent expression patterns of polarization-specific miRNAs led to the identification and demonstrated the regulation of a specific macrophage polarization phenotype, sVEGFR1 by inhibition of miR-125a-5p.


Assuntos
Macrófagos/metabolismo , MicroRNAs/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Células Cultivadas , Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Fator A de Crescimento do Endotélio Vascular/genética
11.
J Gerontol A Biol Sci Med Sci ; 71(8): 992-1004, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26273023

RESUMO

We demonstrated that young male and female mice similarly regenerated injured skeletal muscle; however, female mice transiently increased adipocyte area within regenerated muscle in a sex hormone-dependent manner. We extended these observations to investigate the effect of aging and sex on sarcopenia and muscle regeneration. Cardiotoxin injury to the tibialis anterior muscle of young, middle, and old-aged C57Bl/6J male and female mice was used to measure regenerated myofiber cross-sectional area (CSA), adipocyte area, residual necrosis, and inflammatory cell recruitment. Baseline (uninjured) myofiber CSA was decreased in old mice of both sexes compared to young and middle-aged mice. Regenerated CSA was similar in male mice in all age groups until baseline CSA was attained but decreased in middle and old age female mice compared to young females. Furthermore, adipocyte area within regenerated muscle was transiently increased in young females compared to young males and these sex-dependent increases persisted in middle and old age female mice and were associated with increased Pparg Young female mice had more pro-inflammatory monocytes/macrophages in regenerating muscle than young male mice and increased Sca-1(+)CD45(-)cells. In conclusion, sex and age influence pro-inflammatory cell recruitment, muscle regeneration, and adipocyte area following skeletal muscle injury.


Assuntos
Adipócitos/efeitos dos fármacos , Envelhecimento , Músculo Esquelético/patologia , Regeneração , Sarcopenia/patologia , Animais , Cardiotoxinas/toxicidade , Modelos Animais de Doenças , Feminino , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Músculo Esquelético/lesões , Caracteres Sexuais
12.
PLoS Pathog ; 9(12): e1003857, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24385912

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is causally linked to several human cancers, including Kaposi's sarcoma, primary effusion lymphoma and multicentric Castleman's disease, malignancies commonly found in HIV-infected patients. While KSHV encodes diverse functional products, its mechanism of oncogenesis remains unknown. In this study, we determined the roles KSHV microRNAs (miRs) in cellular transformation and tumorigenesis using a recently developed KSHV-induced cellular transformation system of primary rat mesenchymal precursor cells. A mutant with a cluster of 10 precursor miRs (pre-miRs) deleted failed to transform primary cells, and instead, caused cell cycle arrest and apoptosis. Remarkably, the oncogenicity of the mutant virus was fully restored by genetic complementation with the miR cluster or several individual pre-miRs, which rescued cell cycle progression and inhibited apoptosis in part by redundantly targeting IκBα and the NF-κB pathway. Genomic analysis identified common targets of KSHV miRs in diverse pathways with several cancer-related pathways preferentially targeted. These works define for the first time an essential viral determinant for KSHV-induced oncogenesis and identify NF-κB as a critical pathway targeted by the viral miRs. Our results illustrate a common theme of shared functions with hierarchical order among the KSHV miRs.


Assuntos
Carcinogênese/genética , Proliferação de Células , Transformação Celular Viral/genética , Herpesvirus Humano 8/genética , MicroRNAs/fisiologia , Animais , Sobrevivência Celular/genética , Células Cultivadas , Redes Reguladoras de Genes , Marcação de Genes , Humanos , Análise em Microsséries , Ratos , Transdução de Sinais/genética , Transcriptoma
13.
J Virol ; 86(21): 11698-711, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22915806

RESUMO

Transforming growth factor ß (TGF-ß) signaling regulates cell growth and survival. Dysregulation of the TGF-ß pathway is common in viral infection and cancer. Latent infection by Kaposi's sarcoma-associated herpesvirus (KSHV) is required for the development of several AIDS-related malignancies, including Kaposi's sarcoma and primary effusion lymphoma (PEL). KSHV encodes more than two dozen microRNAs (miRs) derived from 12 pre-miRs with largely unknown functions. In this study, we show that miR variants processed from pre-miR-K10 are expressed in KSHV-infected PEL cells and endothelial cells, while cellular miR-142-3p and its variant miR-142-3p_-1_5, which share the same seed sequence with miR-K10a_ +1_5, are expressed only in PEL cells and not in uninfected and KSHV-infected TIME cells. KSHV miR-K10 variants inhibit TGF-ß signaling by targeting TGF-ß type II receptor (TßRII). Computational and reporter mutagenesis analyses identified three functional target sites in the TßRII 3' untranslated region (3'UTR). Expression of miR-K10 variants is sufficient to inhibit TGF-ß-induced cell apoptosis. A suppressor of the miRs sensitizes latent KSHV-infected PEL cells to TGF-ß and induces apoptosis. These results indicate that miR-K10 variants manipulate the TGF-ß pathway to confer cells with resistance to the growth-inhibitory effect of TGF-ß. Thus, KSHV miRs might target the tumor-suppressive TGF-ß pathway to promote viral latency and contribute to malignant cellular transformation.


Assuntos
Herpesvirus Humano 8/imunologia , Herpesvirus Humano 8/patogenicidade , Evasão da Resposta Imune , MicroRNAs/metabolismo , RNA Viral/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Apoptose , Linhagem Celular , Sobrevivência Celular , Humanos , MicroRNAs/genética , RNA Viral/genética
14.
Virus Genes ; 44(2): 225-36, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22180077

RESUMO

Kaposi's sarcoma-associated herpesvirus ORF30-33 locus encodes four genes with unknown functions. We performed transcriptional mapping of these genes. Northern-hybridization, 5'- and 3'-rapid amplification of cDNA ends, and DNA sequencing identified four transcripts of 3.7, 3.6, 2.7, and 1.4 kb, none of which has alternative splicing. While all transcripts have the same termination site, their start sites vary. All transcripts are not expressed or only weakly expressed in latent cells but can be chemically induced. The 3.7 and 3.6 kb transcripts contain all four genes and are sensitive to cycloheximide (CH) but resistant to phosphonoacetic acid (PAA), indicating that they are early lytic transcripts. The 2.7 kb transcript contains ORF32 and ORF33 genes while the 1.4 kb transcript contains the ORF33 gene. Both transcripts are sensitive to CH and PAA, indicating that they are late lytic transcripts. Furthermore, we identified four promoters with functional TATA boxes, none of which is directly transactivated by RTA. Examination of the 5' untranslated region of ORF31 failed to identify any functional internal ribosome entry sites. These results define the transcriptional patterns of the ORF30-33 locus, which should help the delineation of its function.


Assuntos
Perfilação da Expressão Gênica , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Fases de Leitura Aberta , Transcrição Gênica , Northern Blotting , DNA Complementar/genética , Humanos , Técnicas de Amplificação de Ácido Nucleico , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Sítio de Iniciação de Transcrição
15.
Adv Virol ; 20112011.
Artigo em Inglês | MEDLINE | ID: mdl-21625290

RESUMO

The life cycle of Kaposi's sarcoma-associated herpesvirus (KSHV) consists of latent and lytic replication phases. During latent infection, only a limited number of KSHV genes are expressed. However, this phase of replication is essential for persistent infection, evasion of host immune response, and induction of KSHV-related malignancies. KSHV reactivation from latency produces a wide range of viral products and infectious virions. The resulting de novo infection and viral lytic products modulate diverse cellular pathways and stromal microenvironment, which promote the development of Kaposi's sarcoma (KS). The mechanisms controlling KSHV latency and reactivation are complex, involving both viral and host factors, and are modulated by diverse environmental factors. Here, we review the cellular and molecular basis of KSHV latency and reactivation with a focus on the most recent advancements in the field.

16.
PLoS Pathog ; 7(5): e1002054, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21625536

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) establishes a latent infection in the host following an acute infection. Reactivation from latency contributes to the development of KSHV-induced malignancies, which include Kaposi's sarcoma (KS), the most common cancer in untreated AIDS patients, primary effusion lymphoma and multicentric Castleman's disease. However, the physiological cues that trigger KSHV reactivation remain unclear. Here, we show that the reactive oxygen species (ROS) hydrogen peroxide (H2O2) induces KSHV reactivation from latency through both autocrine and paracrine signaling. Furthermore, KSHV spontaneous lytic replication, and KSHV reactivation from latency induced by oxidative stress, hypoxia, and proinflammatory and proangiogenic cytokines are mediated by H2O2. Mechanistically, H2O2 induction of KSHV reactivation depends on the activation of mitogen-activated protein kinase ERK1/2, JNK, and p38 pathways. Significantly, H2O2 scavengers N-acetyl-L-cysteine (NAC), catalase and glutathione inhibit KSHV lytic replication in culture. In a mouse model of KSHV-induced lymphoma, NAC effectively inhibits KSHV lytic replication and significantly prolongs the lifespan of the mice. These results directly relate KSHV reactivation to oxidative stress and inflammation, which are physiological hallmarks of KS patients. The discovery of this novel mechanism of KSHV reactivation indicates that antioxidants and anti-inflammation drugs could be promising preventive and therapeutic agents for effectively targeting KSHV replication and KSHV-related malignancies.


Assuntos
Herpesvirus Humano 8/fisiologia , Peróxido de Hidrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ativação Viral , Latência Viral , Acetilcisteína/metabolismo , Animais , Western Blotting , Butadienos/farmacologia , Catalase/metabolismo , Linhagem Celular , Imunofluorescência , Glutationa/metabolismo , Células HEK293 , Infecções por Herpesviridae , Herpesvirus Humano 8/metabolismo , Humanos , Imidazóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Estresse Oxidativo , Piridinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma de Kaposi/virologia , Transdução de Sinais , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
J Acquir Immune Defic Syndr ; 56(1): 83-90, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21084997

RESUMO

BACKGROUND: Kaposi's sarcoma-associated herpesvirus (KSHV) seropositivity and lytic antibody titer are predictors for Kaposi's sarcoma. METHODS: We examined demographic, viral, and immunologic factors that influence KSHV latent and lytic antibodies in HIV-infected patients. RESULTS: Detection rate of KSHV latent but not lytic antibodies was lower in patients with CD4 cells/mm3 less than 200 than greater than 200 (odds ratio [OR], 0.26; 95% confidence interval [CI], 0.11-0.61) and CD8 cells/mm3 less than 400 than greater than 400 (OR, 0.26; 95% CI, 0.07-0.67). Overall seropositivity rate was higher in patients with CD4 cells/mm3 less than 200 than greater than 200 (OR, 2.34; 95% CI, 1.37-4.02) and HIV copies/mL greater than 400 than less than 400 (OR, 1.70; 95% CI, 1.09-2.65). Lytic antibody level was inversely correlated with CD4 count (P < 0.001). Lytic seropositivity (OR, 2.47; 95% CI, 1.35-4.50) and antibody level (adjusted difference mean optical density, 0.324; 95% CI, 0.16-0.46) were higher in patients with HIV infection greater than 15 than less than 15 years. Hispanics had higher lytic seropositivity rate (OR, 1.71; 95% CI, 1.07-2.73) and antibody level (adjusted difference mean optical density, 0.111; 95% CI, 0.03-0.18) than non-Hispanics. CONCLUSIONS: Lower CD4 and CD8 counts impair antibody response to KSHV latent antigens. Immune deterioration, long-term HIV infection, and Hispanic status are risk factors for Kaposi's sarcoma predictors.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/imunologia , Fármacos Anti-HIV/uso terapêutico , Anticorpos Antivirais/imunologia , Herpesvirus Humano 8/imunologia , Adulto , Anticorpos Antivirais/sangue , Formação de Anticorpos/imunologia , Antígenos Virais/sangue , Antígenos Virais/imunologia , Terapia Antirretroviral de Alta Atividade , Contagem de Linfócito CD4 , Relação CD4-CD8 , Feminino , Herpesvirus Humano 8/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Fatores de Risco , Sarcoma de Kaposi/imunologia , Sarcoma de Kaposi/virologia , Carga Viral/imunologia , Latência Viral/imunologia
18.
Virulence ; 1(5): 433-5, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21170300

RESUMO

Human herpesviruses have latency and lytic replication phases in their lifecycle. Proper regulation of herpesviral lifecycle is essential for the evasion of host immune surveillance and development of their related diseases. Recent advancements indicate a role of a novel class of viral non-coding RNAs, microRNA (miRNA), in the fine-tuning of herpesviral lifecycle. So far, herpesviral miRNAs appear to promote viral latency by inhibiting viral lytic replication either through direct targeting of key viral replication genes or through manipulation of host pathways that regulate viral lifecycle. The oncogenic Kaposi sarcoma-associated herpesvirus (KSHV) has adapted both strategies to control viral latency. Our recent study has identified a KSHV miRNA that inhibits viral lytic replication by upregulating the NFκB pathway.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/fisiologia , MicroRNAs/metabolismo , RNA Viral/metabolismo , Latência Viral , Herpesvirus Humano 8/genética , Humanos , MicroRNAs/genética , Modelos Biológicos , NF-kappa B/biossíntese , RNA Viral/genética , Regulação para Cima
20.
Nat Cell Biol ; 12(2): 193-9, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20081837

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is causally linked to several acquired immune deficiency syndrome-related malignancies, including Kaposi's sarcoma, primary effusion lymphoma (PEL) and a subset of multicentric Castleman's disease. Control of viral lytic replication is essential for KSHV latency, evasion of the host immune system and induction of tumours. Here, we show that deletion of a 14 microRNA (miRNA) cluster from the KSHV genome significantly enhances viral lytic replication as a result of reduced NF-kappaB activity. The miRNA cluster regulates the NF-kappaB pathway by reducing expression of IkappaBalpha protein, an inhibitor of NF-kappaB complexes. Computational and miRNA seed mutagenesis analyses were used to identify KSHV miR-K1, which directly regulates the IkappaBalpha protein level by targeting the 3'UTR of its transcript. Expression of miR-K1 is sufficient to rescue NF-kappaB activity and inhibit viral lytic replication, whereas inhibition of miR-K1 in KSHV-infected PEL cells has the opposite effect. Thus, KSHV encodes an miRNA to control viral replication by activating the NF-kappaB pathway. These results demonstrate an important role for KSHV miRNAs in regulating viral latency and lytic replication by manipulating the host survival pathway.


Assuntos
Herpesvirus Humano 8/crescimento & desenvolvimento , Herpesvirus Humano 8/metabolismo , Proteínas I-kappa B/metabolismo , MicroRNAs/metabolismo , Replicação Viral/fisiologia , Northern Blotting , Western Blotting , Linhagem Celular , Ensaio de Desvio de Mobilidade Eletroforética , Imunofluorescência , Herpesvirus Humano 8/genética , Humanos , Proteínas I-kappa B/genética , MicroRNAs/genética , Inibidor de NF-kappaB alfa , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA