Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Immunol ; 21(8): 950-961, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32572241

RESUMO

A contribution of epigenetic modifications to B cell tolerance has been proposed but not directly tested. Here we report that deficiency of ten-eleven translocation (Tet) DNA demethylase family members Tet2 and Tet3 in B cells led to hyperactivation of B and T cells, autoantibody production and lupus-like disease in mice. Mechanistically, in the absence of Tet2 and Tet3, downregulation of CD86, which normally occurs following chronic exposure of self-reactive B cells to self-antigen, did not take place. The importance of dysregulated CD86 expression in Tet2- and Tet3-deficient B cells was further demonstrated by the restriction, albeit not complete, on aberrant T and B cell activation following anti-CD86 blockade. Tet2- and Tet3-deficient B cells had decreased accumulation of histone deacetylase 1 (HDAC1) and HDAC2 at the Cd86 locus. Thus, our findings suggest that Tet2- and Tet3-mediated chromatin modification participates in repression of CD86 on chronically stimulated self-reactive B cells, which contributes, at least in part, to preventing autoimmunity.


Assuntos
Autoimunidade/imunologia , Linfócitos B/imunologia , Antígeno B7-2/imunologia , Proteínas de Ligação a DNA/imunologia , Dioxigenases/imunologia , Proteínas Proto-Oncogênicas/imunologia , Animais , Doenças Autoimunes/imunologia , Epigênese Genética/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
2.
J Allergy Clin Immunol ; 143(3): 1163-1175.e15, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30053529

RESUMO

BACKGROUND: Chronic rhinosinusitis with nasal polyposis (CRSwNP) is characterized by eosinophilic inflammation and polyposis at the nose and paranasal sinus and a high concentration of IgE in nasal polyps (NPs). The causative antigen and pathogenesis of CRSwNP remain unknown. OBJECTIVE: We aimed to identify reactive allergens of IgE antibodies produced locally in NPs of patients with CRSwNP. We also attempted to unravel the differentiation pathway of IgE-producing B cells in NPs. METHODS: IgE reactivity of patients with CRSwNP was investigated by characterizing single cell-derived mAbs. T-cell response against identified allergens was investigated in vitro. NP-infiltrating lymphocytes were characterized by using flow cytometry. Immunoglobulins expressed in NPs were analyzed by using high-throughput DNA sequencing for immunoglobulin. RESULTS: About 20% of isolated IgE antibodies derived from NP-residing plasmablasts specifically recognized surface determinants of nasal bacteria, such as Staphylococcus aureus, Streptococcus pyogenes, and Haemophilus influenzae. A TH2 response against S pyogenes was observed in patients with CRSwNP. Flow cytometric analysis revealed sizable germinal center B-like cell and plasmablast subsets expressing IgE on the cell surface in NPs. High-throughput DNA sequencing immunoglobulin analysis highlighted the clonal connectivity of IgE with IgG and IgA1. The Iε-Cα1 circle transcript was detected in NPs. CONCLUSIONS: In patients with CRSwNP, nasal bacteria-reactive B cells differentiate into IgE-producing B cells through IgG/IgA1-IgE class switching, suggesting that allergic conversion of the mucosal response against nasal bacteria underlies disease pathogenesis.


Assuntos
Linfócitos B/imunologia , Bactérias/imunologia , Imunidade nas Mucosas , Imunoglobulina E/imunologia , Pólipos Nasais/imunologia , Rinite/imunologia , Sinusite/imunologia , Adulto , Idoso , Anticorpos Monoclonais/farmacologia , Células Cultivadas , Doença Crônica , Eosinofilia/imunologia , Eosinofilia/microbiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/imunologia , Mucosa Nasal/microbiologia , Pólipos Nasais/microbiologia , Rinite/microbiologia , Sinusite/microbiologia , Adulto Jovem
3.
Int Immunol ; 30(12): 579-590, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30137504

RESUMO

Epstein-Barr virus (EBV)-encoded latent membrane protein 1 (LMP1), which mimics a constitutively active receptor, is required for viral transformation of primary B cells. LMP1 is expressed in EBV-infected germinal center (GC) B cells of immunocompetent individuals, suggesting that it may contribute to persistent EBV infection. In this study, we generated and analyzed mice that expressed LMP1 under the control of the CD19 or activation-induced cytidine deaminase (AID) promoter. Expression of LMP1 induced activation of B cells but severely inhibited their differentiation into antibody-secreting cells (ASCs) in vitro and GC B cells in vivo. LMP1-expressing (LMP1+) B cells not only suppressed the functions of wild-type (WT) B cells in in vitro co-culture, but also blocked differentiation of WT B cells into GC B cells and ASCs in immunized bone marrow chimeric mice. Microarray analysis revealed that the gene encoding indoleamine 2,3-dioxygenase 1 (IDO1), a major enzyme involved in the tryptophan metabolic process, was highly induced by LMP1. Either inhibition of IDO1 activity by methyl-l-tryptophan or knockout of Ido1 in LMP1+ B cells could rescue WT B cells from such suppression. IDO1-induced tryptophan consumption and production of tryptophan metabolites appeared to be responsible for inhibition of B-cell function. We conclude that LMP1 expression in antigen-committed B cells not only directly impairs GC B-cell differentiation, but also indirectly inhibits the functions of neighboring B cells, resulting in suppression of humoral immune responses. Such bystander inhibition by LMP1+ B cells may contribute to immune evasion by EBV.


Assuntos
Linfócitos B/imunologia , Herpesvirus Humano 4/imunologia , Imunidade Humoral/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Diferenciação Celular , Camundongos , Camundongos Transgênicos
4.
Sci Rep ; 7(1): 16428, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29180749

RESUMO

The evolutional process of disease-associated autoantibodies in systemic lupus erythematosus (SLE) remains to be established. Here we show intraclonal diversification and affinity maturation of anti-nuclear antibody (ANA)-producing B cells in SLE. We identified a panel of monoclonal ANAs recognizing nuclear antigens, such as double-stranded DNA (dsDNA) and ribonucleoproteins (RNPs) from acute SLE subjects. These ANAs had relatively few, but nonetheless critical mutations. High-throughput immunoglobulin sequencing of blood lymphocytes disclosed the existence of sizable ANA lineages shearing critical mutations intraclonally. We further focused on anti-DNA antibodies, which are capable to bind to both single-stranded (ss) and dsDNA at high affinity. Crystal structure and biochemical analysis confirmed a direct role of the mutations in the acquisition of DNA reactivity and also revealed that these anti-DNA antibodies recognized an unpaired region within DNA duplex. Our study unveils the unique properties of high-affinity anti-DNA antibodies that are generated through antigen-driven affinity maturation in acute phase of SLE.


Assuntos
Anticorpos Antinucleares/imunologia , Antígenos/imunologia , Evolução Clonal/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Doença Aguda , Sequência de Aminoácidos , Anticorpos Antinucleares/química , Antígenos/química , Autoanticorpos/sangue , DNA/imunologia , Células HEK293 , Humanos , Lúpus Eritematoso Sistêmico/sangue , Mutação/genética , Taxa de Mutação , Filogenia , Sindecana-1/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(37): 11612-7, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26305967

RESUMO

Epstein-Barr virus (EBV) infects germinal center (GC) B cells and establishes persistent infection in memory B cells. EBV-infected B cells can cause B-cell malignancies in humans with T- or natural killer-cell deficiency. We now find that EBV-encoded latent membrane protein 2A (LMP2A) mimics B-cell antigen receptor (BCR) signaling in murine GC B cells, causing altered humoral immune responses and autoimmune diseases. Investigation of the impact of LMP2A on B-cell differentiation in mice that conditionally express LMP2A in GC B cells or all B-lineage cells found LMP2A expression enhanced not only BCR signals but also plasma cell differentiation in vitro and in vivo. Conditional LMP2A expression in GC B cells resulted in preferential selection of low-affinity antibody-producing B cells despite apparently normal GC formation. GC B-cell-specific LMP2A expression led to systemic lupus erythematosus-like autoimmune phenotypes in an age-dependent manner. Epigenetic profiling of LMP2A B cells found increased H3K27ac and H3K4me1 signals at the zinc finger and bric-a-brac, tramtrack domain-containing protein 20 locus. We conclude that LMP2A reduces the stringency of GC B-cell selection and may contribute to persistent EBV infection and pathogenesis by providing GC B cells with excessive prosurvival effects.


Assuntos
Centro Germinativo/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Autoanticorpos/química , Doenças Autoimunes/metabolismo , Doenças Autoimunes/virologia , Diferenciação Celular , Linhagem da Célula , Cruzamentos Genéticos , Epigênese Genética , Citometria de Fluxo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Heterozigoto , Imunidade Humoral , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais , Baço/citologia , Dedos de Zinco
6.
Viruses ; 6(9): 3472-86, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25243371

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV, also named Human herpesvirus 8 HHV-8) is the cause of Kaposi sarcoma (KS), the most common malignancy in HIV-infected individuals worldwide, primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). KSHV is a double-stranded DNA virus that encodes several homologues of cellular proteins. The structural similarity between viral and host proteins explains why some viral homologues function as their host counterparts, but sometimes at unusual anatomical sites and inappropriate times. In other cases, structural modification in the viral proteins can suppress or override the function of the host homologue, contributing to KSHV-related diseases. For example, viral IL-6 (vIL-6) is sufficiently different from human IL-6 to activate gp130 signaling independent of the α subunit. As a consequence, vIL-6 can activate many cell types that are unresponsive to cellular IL-6, contributing to MCD disease manifestations. Here, we discuss the molecular biology of KSHV homologues of cellular products as conduits of virus/host interaction with a focus on identifying new strategies for therapy of KS and other KSHV-related diseases.


Assuntos
Hiperplasia do Linfonodo Gigante/etiologia , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Linfoma de Efusão Primária/etiologia , Sarcoma de Kaposi/etiologia , Citocinas/genética , Citocinas/metabolismo , Infecções por Herpesviridae/genética , Humanos , Mediadores da Inflamação/metabolismo , NF-kappa B/metabolismo , Proteína Supressora de Tumor p53/metabolismo
7.
Haematologica ; 97(6): 818-26, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22271895

RESUMO

BACKGROUND: Mobilization of hematopoietic stem/progenitor cells from the bone marrow to the peripheral blood by granulocyte colony-stimulating factor is the primary means to acquire stem cell grafts for hematopoietic cell transplantation. Since hematopoietic stem/progenitor cells represent a minority of all blood cells mobilized by granulocyte colony-stimulating factor, the underlying mechanisms need to be understood in order to develop selective drugs. DESIGN AND METHODS: We analyzed phenotypic, biochemical and genetic changes in bone marrow cell populations from granulocyte colony-stimulating factor-mobilized and control mice, and linked such changes to effective mobilization of hematopoietic stem/progenitor cells. RESULTS: We show that granulocyte colony-stimulating factor indirectly reduces expression of surface vascular cell adhesion molecule 1 on bone marrow hematopoietic stem/progenitor cells, stromal cells and endothelial cells by promoting the accumulation of microRNA-126 (miR126)-containing microvescicles in the bone marrow extracellular compartment. We found that hematopoietic stem/progenitor cells, stromal cells and endothelial cells readily incorporate these miR126-loaded microvescicles, and that miR126 represses vascular cell adhesion molecule 1 expression on bone marrow hematopoietic stem/progenitor cells, stromal cells and endothelial cells. In line with this, miR126-null mice displayed a reduced mobilization response to granulocyte colony-stimulating factor. CONCLUSIONS: Our results implicate miR126 in the regulation of hematopoietic stem/progenitor cell trafficking between the bone marrow and peripheral sites, clarify the role of vascular cell adhesion molecule 1 in granulocyte colony-stimulating factor-mediated mobilization, and have important implications for improved approaches to selective mobilization of hematopoietic stem/progenitor cells.


Assuntos
Medula Óssea/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , MicroRNAs/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Animais , Medula Óssea/imunologia , Medula Óssea/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Vesículas Citoplasmáticas/imunologia , Vesículas Citoplasmáticas/metabolismo , Regulação para Baixo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Espaço Extracelular/imunologia , Espaço Extracelular/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia
8.
J Interferon Cytokine Res ; 31(11): 791-801, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21767154

RESUMO

Viral interleukin-6 (vIL-6) is a product of Kaposi's sarcoma-associated herpesvirus (KSHV) expressed in latently infected cells and to a higher degree during viral replication. A distinctive feature of vIL-6 is the ability to directly bind and activate gp130 signaling in the absence of other receptor subunits. Secretion of vIL-6 is generally poor, but vIL-6 can activate gp130 from inside the cell. Due to the wide cell distribution of gp130, vIL-6 has the potential to induce a wide range of biological effects. Expression of vIL-6 is variable in KSHV-associated Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), multicentric Castleman's disease (MCD), and in a newly described MCD-like systemic inflammatory syndrome observed in human immunodeficiency virus-positive patients. PEL effusions usually contain vIL-6 at high concentrations; since vIL-6 induces vascular endothelial growth factor, vIL-6 likely contributes to vascular permeability and formation of PEL effusions. Lymph nodes affected with MCD contain vIL-6-positive cells, and vIL-6 levels rise in conjunction with flares of the disease and likely contribute to symptoms of inflammation. The development of vIL-6 inhibitors is a potentially important advance in the treatment of KSHV-associated malignancies where vIL-6 is expressed.


Assuntos
Hiperplasia do Linfonodo Gigante/metabolismo , Herpesvirus Humano 8/metabolismo , Interleucina-6/metabolismo , Linfoma de Efusão Primária/metabolismo , Sarcoma de Kaposi/metabolismo , Animais , Hiperplasia do Linfonodo Gigante/patologia , Hiperplasia do Linfonodo Gigante/virologia , Herpesvirus Humano 8/genética , Humanos , Interleucina-6/genética , Linfoma de Efusão Primária/patologia , Linfoma de Efusão Primária/virologia , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia
9.
J Virol ; 85(7): 3179-86, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21270144

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is a gammaretrovirus linked to prostate carcinoma and chronic fatigue syndrome. Here we report that NF-κB activation can markedly increase XMRV production. The inflammatory cytokine tumor necrosis factor alpha (TNF-α), which activates NF-κB, significantly augmented viral Gag protein production in XMRV-infected cells. Reporter assays showed that TNF-α and Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1), an intrinsic NF-κB activator, increased long terminal repeat (LTR)-dependent XMRV transcription. We identified two NF-κB binding sites (designated κB-1 and κB-2) in the LTR U3 region of XMRV and demonstrated that both sites bind to the NF-κB component p65/RelA. Mutation of the κB-1 site, but not the κB-2 site, impaired responsiveness to TNF-α and LMP1 in reporter assays. A mutant XMRV with a mutation at the κB-1 site replicated significantly less efficiently than the wild-type XMRV in the prostate carcinoma LNCaP, DU145, and PC-3 cell lines, HEK293 cells, the EBV-immortalized cell line IB4, and the Burkitt's lymphoma cell line BJAB. These results demonstrate that TNF-α and EBV LMP1 enhance XMRV replication in prostate carcinoma and B-lineage cells through the κB-1 site in the XMRV LTR, suggesting that inflammation, EBV infection, and other conditions leading to NF-κB activation may promote XMRV spread in humans.


Assuntos
Linfócitos B/virologia , Carcinoma/virologia , NF-kappa B/metabolismo , Neoplasias da Próstata/virologia , Transcrição Gênica , Replicação Viral , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/fisiologia , Sítios de Ligação , Linhagem Celular Tumoral , Produtos do Gene gag/biossíntese , Humanos , Masculino , Ligação Proteica , Sequências Repetidas Terminais/genética , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Blood ; 115(19): 3970-9, 2010 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-20203268

RESUMO

The transcription factor growth factor independence 1 (Gfi1) and the growth factor granulocyte colony-stimulating factor (G-CSF) are individually essential for neutrophil differentiation from myeloid progenitors. Here, we provide evidence that the functions of Gfi1 and G-CSF are linked in the regulation of granulopoiesis. We report that Gfi1 promotes the expression of Ras guanine nucleotide releasing protein 1 (RasGRP1), an exchange factor that activates Ras, and that RasGRP1 is required for G-CSF signaling through the Ras/mitogen-activated protein/extracellular signal-regulated kinase (MEK/Erk) pathway. Gfi1-null mice have reduced levels of RasGRP1 mRNA and protein in thymus, spleen, and bone marrow, and Gfi1 transduction in myeloid cells promotes RasGRP1 expression. When stimulated with G-CSF, Gfi1-null myeloid cells are selectively defective at activating Erk1/2, but not signal transducer and activator of transcription 1 (STAT1) or STAT3, and fail to differentiate into neutrophils. Expression of RasGRP1 in Gfi1-deficient cells rescues Erk1/2 activation by G-CSF and allows neutrophil maturation by G-CSF. These results uncover a previously unknown function of Gfi1 as a regulator of RasGRP1 and link Gfi1 transcriptional control to G-CSF signaling and regulation of granulopoiesis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Transdução de Sinais , Fatores de Transcrição/fisiologia , Animais , Western Blotting , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Células-Tronco Hematopoéticas/metabolismo , Técnicas Imunoenzimáticas , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/metabolismo
11.
J Virol ; 83(23): 12368-77, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19776126

RESUMO

B lymphocytes converted into lymphoblastoid cell lines (LCLs) by an Epstein-Barr virus that expresses a conditional EBNA3C require complementation with EBNA3C for growth under nonpermissive conditions. Complementation with relatively large EBNA3C deletion mutants identified amino acids (aa) 1 to 506 (which includes the RBP-Jkappa/CSL [RBP-Jkappa] binding domain) and 733 to 909 to be essential for LCL growth, aa 728 to 732 and 910 to 992 to be important for full wild-type (wt) growth, and only aa 507 to 727 to be unimportant (S. Maruo, Y. Wu, T. Ito, T. Kanda, E. D. Kieff, and K. Takada, Proc. Natl. Acad. Sci. USA 106:4419-4424, 2009). When mutants with smaller deletions were used, only aa 51 to 400 and 851 to 900 were essential for LCL growth; aa 447 to 544, 701 to 750, 801 to 850, and 901 to 992 were important for full wt growth; and aa 4 to 50, 401 to 450, 550 to 707, and 751 to 800 were unimportant. These data reduce the EBNA3C essential residues from 68% to 40% of the open reading frame. Point mutations confirmed RBP-Jkappa binding to be essential for wt growth and indicated that SUMO and CtBP binding interactions were important only for full wt growth. EBNA3C aa 51 to 150, 249 to 311, and 851 to 900 were necessary for maintaining LCL growth, but not RBP-Jkappa interaction, and likely mediate interactions with other key cell proteins. Moreover, all mutants null for LCL growth had fewer S+G(2)/M-phase cells at 14 days, consistent with EBNA3C interaction with RBP-Jkappa as well as aa 51 to 150, 249 to 311, and 851 to 900 being required to suppress p16(INK4A) (S. Maruo, Y. Wu, S. Ishikawa, T. Kanda, D. Iwakiri, and K. Takada, Proc. Natl. Acad. Sci. USA 103:19500-19505, 2006). We have confirmed that EBNA3C upregulates TCL1 and discovered that EBNA3C upregulates TCL1 through RBP-Jkappa, indicating a central role for EBNA3C interaction with RBP-Jkappa in mediating cell gene transcription.


Assuntos
Antígenos Virais/fisiologia , Transformação Celular Neoplásica , Herpesvirus Humano 4/patogenicidade , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Antígenos Virais/genética , Linhagem Celular Tumoral , Antígenos Nucleares do Vírus Epstein-Barr , Herpesvirus Humano 4/genética , Humanos , Mutagênese Sítio-Dirigida , Mutação Puntual , Domínios e Motivos de Interação entre Proteínas , Deleção de Sequência , Regulação para Cima
12.
Future Microbiol ; 4(7): 903-17, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19722843

RESUMO

Tumor angiogenesis is the process by which new blood vessels are formed within emerging or progressing malignancies. The human Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus critically contribute to the pathogenesis of selected tumor types, including nasopharyngeal carcinoma and Kaposi's sarcoma, respectively, where angiogenesis is robust and often disrupted. Lymphangiogenesis, the process by which new lymphatic vessels are formed, is also induced in Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus-associated malignancies and in some cases may contribute to metastasis. Recent studies have identified a number of molecules and signaling pathways that underlie angiogenesis and lymphangiogenesis, and clarified the pivotal role of the VEGF family of proteins and their receptors. New treatment modalities that target members of this family have gained approval for clinical use in cancer. Pathogenetic steps are often difficult to dissect in many cancer types, but virus-induced malignancies provide a unique opportunity for understanding the molecular regulation of cancer progression, including angiogenesis. Dissection of viral gene contribution to tumor angiogenesis could result in a better understanding of the angiogenic process, its contribution to cancer and help in the design of rational therapies that target tumor growth and vascularization.


Assuntos
Infecções por Vírus Epstein-Barr/patologia , Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 8/patogenicidade , Neoplasias Nasofaríngeas/patologia , Neovascularização Patológica/virologia , Sarcoma de Kaposi/patologia , Humanos , Modelos Biológicos , Neoplasias Nasofaríngeas/virologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Sarcoma de Kaposi/virologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
13.
Blood ; 114(8): 1707-16, 2009 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-19411631

RESUMO

EphrinB transmembrane ligands and their cognate EphB receptor tyrosine kinases regulate vascular development through bidirectional cell-to-cell signaling, but little is known about the role of EphrinB during postnatal vascular remodeling. We report that EphrinB is a critical mediator of postnatal pericyte-to-endothelial cell assembly into vascular structures. This function is dependent upon extracellular matrix-supported cell-to-cell contact, engagement of EphrinB by EphB receptors expressed on another cell, and Src-dependent phosphorylation of the intracytoplasmic domain of EphrinB. Phosphorylated EphrinB marks angiogenic blood vessels in the developing and hypoxic retina, the wounded skin, and tumor tissue, and is detected at contact points between endothelial cells and pericytes. Furthermore, inhibition ofEphrinB activity prevents proper assembly of pericytes and endothelial cells into vascular structures. These results reveal a role for EphrinB signaling in orchestrating pericyte/endothelial cell assembly, and suggest that therapeutic targeting of EphrinB may prove useful for disrupting angiogenesis when it contributes to disease.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Células Endoteliais/fisiologia , Efrinas/fisiologia , Neovascularização Fisiológica/fisiologia , Pericitos/fisiologia , Animais , Animais Recém-Nascidos , Vasos Sanguíneos/metabolismo , Células da Medula Óssea/fisiologia , Adesão Celular/genética , Células Cultivadas , Células Endoteliais/metabolismo , Efrina-B2/antagonistas & inibidores , Efrina-B2/genética , Efrina-B2/fisiologia , Efrinas/genética , Efrinas/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Neovascularização Fisiológica/genética , Pericitos/metabolismo , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo , Receptores da Família Eph/fisiologia , Transdução de Sinais/fisiologia
14.
J Virol ; 83(5): 2140-53, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19091861

RESUMO

Kaposi's sarcoma (KS) is an angioproliferative inflammatory disorder induced by endothelial cell infection with the KS-associated herpesvirus (KSHV). ORFK13/vFLIP, one of the KSHV genes expressed in KS, encodes a 188-amino-acid protein which binds to the Ikappab kinase (IKK) complex to activate NF-kappaB. We examined ORFK13/vFLIP contribution to KS phenotype and potential for therapeutic targeting. Retroviral transduction of ORFK13/vFLIP into primary human endothelial cells induces the spindle morphology distinctive of KS cells and promotes the formation of abnormal vascular networks typical of KS vasculature; upregulates the expression of proinflammatory cytokines, chemokines, and interferon-responsive genes; and stimulates the adhesion of inflammatory cells characteristic of KS lesions. Thymidine phosphorylase, a cellular enzyme markedly induced by ORFK13/vFLIP, can metabolize the prodrug 5-fluoro-5-deoxyuridine (5-dFUrd) to 5-fluouridine (5-FU), a potent thymidine synthase inhibitor, which blocks DNA and RNA synthesis. When tested for cytotoxicity, 5-dFUrd (0.1 to 1 microM) selectively killed ORFK13/vFLIP-expressing endothelial cells while sparing control cells. These results demonstrate that ORFK13/vFLIP directly and indirectly contributes to the inflammatory and vascular phenotype of KS and identify 5-dFUrd as a potential new drug that targets KSHV latency for the treatment of KS and other KSHV-associated malignancies.


Assuntos
Células Endoteliais/virologia , Herpesvirus Humano 8/metabolismo , Sarcoma de Kaposi/genética , Proteínas Virais/metabolismo , Forma Celular , Células Cultivadas , Células Endoteliais/citologia , Regulação da Expressão Gênica , Herpesvirus Humano 8/genética , Humanos , Sarcoma de Kaposi/virologia , Transdução Genética , Proteínas Virais/genética
15.
J Leukoc Biol ; 84(4): 994-1000, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18319288

RESUMO

Kaposi's sarcoma (KS)-associated herpesvirus is associated with the proliferative/malignant disorders KS, primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD) in patients with AIDS. In spite of recent advances in the treatment of KS, PEL and MCD represent therapeutic challenges. Recent advances in dissecting the pathogenesis of these diseases have indicated that the viral cytokine IL-6 and the cellular cytokines/growth factors IL-10, IL-6, stromal cell-derived factor 1, and vascular endothelial growth factor are important contributors to the growth, survival, and spread of PEL and MCD and are therefore potential targets for drug development.


Assuntos
Citocinas/fisiologia , Herpesvirus Humano 8/imunologia , Sarcoma de Kaposi/imunologia , Síndrome da Imunodeficiência Adquirida/complicações , Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Animais , Terapia Antirretroviral de Alta Atividade , Divisão Celular , Sobrevivência Celular , Modelos Animais de Doenças , Herpesvirus Humano 8/genética , Humanos , Interleucina-10/imunologia , Interleucina-6/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Sarcoma de Kaposi/complicações , Sarcoma de Kaposi/patologia , Fator A de Crescimento do Endotélio Vascular/imunologia
16.
Virus Res ; 122(1-2): 85-94, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16920214

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is a causative agent for some tumors. The virus establishes latency in infected cells, where its genomes are often present as episomes and replicate in a cell-cycle-dependent manner, apparently maintaining the same copy number. LANA and TR are key KSHV replication factors, and we hypothesized that they also function in viral genome maintenance. We cloned a bacmid containing the viral TR region from PEL cells and tested whether TR with LANA were sufficient for viral genome maintenance. However, neither the TR region nor even the full KSHV genome cloned into a bacmid were maintained in cultured cells, except when they were grown under selective pressure. Thus, no specific viral mechanism for the faithful partitioning and maintenance of the KSHV genome is likely to exist. KSHV might confer a positive growth effect on infected PEL cells, but not on immortalized or transformed cells previously uninfected by KSHV.


Assuntos
DNA Viral/metabolismo , Genoma Viral , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Replicação Viral , Antígenos Virais/fisiologia , Linhagem Celular , Clonagem Molecular , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Microscopia de Fluorescência , Proteínas Nucleares/fisiologia
17.
Biochem Biophys Res Commun ; 345(1): 410-8, 2006 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-16678790

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is a gamma-herpesvirus that is frequently associated with the formation of tumors like Kaposi's sarcoma and primary effusion lymphomas. KSHV RTA is required for lytic replication/reactivation in KSHV-infected cells. We developed an inducible RTA expression cell line to search for RTA-inducible genes. Using DNA chip technology, we showed that hey1, a basic helix-loop-helix-containing transcription factor, increased three- to fourfold in this line. In a luciferase reporter system, RTA activated the hey1 promoter in several cell lines; conversely, HEY1 repressed the rta promoter. Chromatin immunoprecipitation assays indicated that HEY did not bind the rta promoter directly, although it may repress rta expression as part of a complex. Thus, hey1 may help to downregulate RTA after its burst of expression during lytic replication/reactivation. Similar repression mechanisms may downregulate immediate-early gene expression in other herpesviruses, although different transcription factors are probably involved.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Ciclo Celular/genética , Genes Precoces/genética , Transativadores/genética , Ativação Transcricional/genética , Animais , Células COS , Chlorocebus aethiops , Humanos , Camundongos , Regiões Promotoras Genéticas/genética
18.
J Virol ; 78(18): 9936-46, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15331727

RESUMO

During latency, Kaposi's sarcoma-associated herpesvirus (KSHV) is thought to replicate once and to be partitioned in synchrony with the cell cycle of the host. In this replication cycle, the KSHV terminal repeat (TR) sequence functions as a replication origin, assisted by the latency-associated nuclear antigen (LANA). Thus, TR seems to function as a cis element for the replication and partitioning of the KSHV genome. Viral replication and partitioning are also likely to require cellular factors that interact with TR in either a LANA-dependent or -independent manner. Here, we sought to identify factors that associate with TR by using a TR DNA column and found that poly(ADP-ribose) polymerase 1 (PARP1) and known replication factors, including ORC2, CDC6, and Mcm7, bound to TR. PARP1 bound directly to a specific region within TR independent of LANA, and LANA was poly(ADP-ribosyl)ated by PARP1. Drugs such as hydroxyurea and niacinamide, which raise or lower PARP activity, respectively, affected the virus copy number in infected cells. Thus, the poly(ADP-ribosyl)ation status of LANA appears to affect the replication and/or maintenance of the viral genome. Drugs that specifically up-regulate PARP activity may lead to the disappearance of latent KSHV.


Assuntos
DNA Viral/genética , DNA Viral/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Antígenos Virais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular Tumoral , Genoma Viral , Herpesvirus Humano 8/patogenicidade , Humanos , Proteínas Nucleares/metabolismo , Origem de Replicação , Sequências Repetidas Terminais , Latência Viral , Replicação Viral
19.
Virology ; 325(2): 364-78, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15246275

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV; also known as human herpesvirus 8, HHV-8) belongs to the gamma-herpesvirus subfamily. The KSHV ORF57 gene is thought to be a homolog of posttranscriptional regulators that are conserved in the herpesvirus family and are essential for replication. We generated specific monoclonal antibodies (mAbs) against the ORF57 protein that detected the 51-kDa protein expressed in the nucleus of KSHV-infected cells. We also found that the ORF57 protein interacted with poly(rC)-binding protein 1 (PCBP1), a cellular RNA-binding, posttranscriptional regulator. ORF57's interaction with PCBP1 enhanced the activity of not only poliovirus internal ribosome-entry site (IRES)-dependent translation but also X-linked inhibitor of apoptosis (XIAP) and KSHV vFLIP IRES. Actually, when ORF57 expression was induced by the expression of replication and transcription activator (RTA) in KSHV-infected cells, the expression of XIAP was enhanced. These results suggest that ORF57 binds to PCBP1 as a functional partner for posttranscriptional regulation and is involved in the regulation of the expression of both cellular and viral genes through IRESs.


Assuntos
Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , DNA Complementar/genética , DNA Viral/genética , Proteínas de Ligação a DNA , Regulação Viral da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Mapeamento de Peptídeos , Processamento Pós-Transcricional do RNA , RNA Viral/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA , Proteínas Virais/fisiologia
20.
J Virol ; 78(14): 7299-310, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15220403

RESUMO

In the latent infection of Kaposi's sarcoma-associated herpesvirus (KSHV), its 160-kb circularized episomal DNA is replicated and maintained in the host nucleus. KSHV latency-associated nuclear antigen (LANA) is a key factor for maintaining viral latency. LANA binds to the terminal repeat (TR) DNA of the viral genome, leading to its localization to specific dot structures in the nucleus. In such an infected cell, the expression of the viral genes is restricted by a mechanism that is still unclear. Here, we found that LANA interacts with SUV39H1 histone methyltransferase, a key component of heterochromatin formation, as determined by use of a DNA pull-down assay with a biotinylated DNA fragment that contained a LANA-specific binding sequence and a maltose-binding protein pull-down assay. The diffuse localization of LANA on the chromosomes of uninfected cells changed to a punctate one with the introduction of a bacterial artificial chromosome containing most of the TR region, and SUV39H1 clearly colocalized with the LANA-associated dots. Thus, the LANA foci in KSHV-infected cells seemed to include SUV39H1 as well as heterochromatin protein 1. Furthermore, a chromatin immunoprecipitation assay revealed that the TR and the open reading frame (ORF) K1 and ORF50/RTA genes, but not the ORF73/LANA gene, lay within the heterochromatin during KSHV latency. Taken together, these observations indicate that LANA recruits heterochromatin components to the viral genome, which may lead to the establishment of viral latency and govern the transcription program.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/fisiologia , Heterocromatina/metabolismo , Proteínas Nucleares/metabolismo , Transcrição Gênica , Latência Viral , Antígenos Virais , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Herpesvirus Humano 8/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Metiltransferases/metabolismo , Dados de Sequência Molecular , Proteínas Repressoras/metabolismo , Sequências Repetidas Terminais/genética , Transativadores/genética , Transativadores/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA