Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Front Immunol ; 13: 959962, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189216

RESUMEN

HTLV-1 is an oncovirus causing ATL and other inflammatory diseases such as HAM/TSP and HU in about 5% of infected individuals. It is also known that HTLV-1-infected cells maintain a disease-free, immortalized, latent state throughout the lifetimes of about 95% of infected individuals. We believe that the stable maintenance of disease-free infected cells in the carrier is an intrinsic characteristic of HTLV-1 that has been acquired during its evolution in the human life cycle. We speculate that the pathogenesis of the virus is ruled by the orchestrated functions of viral proteins. In particular, the regulation of Rex, the conductor of viral replication rate, is expected to be closely related to the viral program in the early active viral replication followed by the stable latency in HTLV-1 infected T cells. HTLV-1 and HIV-1 belong to the family Retroviridae and share the same tropism, e.g., human CD4+ T cells. These viruses show significant similarities in the viral genomic structure and the molecular mechanism of the replication cycle. However, HTLV-1 and HIV-1 infected T cells show different phenotypes, especially in the level of virion production. We speculate that how the activity of HTLV-1 Rex and its counterpart HIV-1 Rev are regulated may be closely related to the properties of respective infected T cells. In this review, we compare various pathological aspects of HTLV-1 and HIV-1. In particular, we investigated the presence or absence of a virally encoded "regulatory valve" for HTLV-1 Rex or HIV-1 Rev to explore its importance in the regulation of viral particle production in infected T cells. Finally, wereaffirm Rex as the key conductor for viral replication and viral pathogenesis based on our recent study on the novel functional aspects of Rex. Since the activity of Rex is closely related to the viral replication rate, we hypothesize that the "regulatory valve" on the Rex activity may have been selectively evolved to achieve the "scenario" with early viral particle production and the subsequent long, stable deep latency in HTLV-1 infected cells.


Asunto(s)
VIH-1 , Virus Linfotrópico T Tipo 1 Humano , Productos del Gen rex/genética , Productos del Gen rex/metabolismo , Productos del Gen tax/genética , Productos del Gen tax/metabolismo , VIH-1/genética , Virus Linfotrópico T Tipo 1 Humano/fisiología , Humanos , Proteínas Virales/metabolismo , Replicación Viral
2.
Viruses ; 14(2)2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35215946

RESUMEN

The human retrovirus human T-cell leukemia virus type I (HTLV-1) infects human T cells by vertical transmission from mother to child through breast milk or horizontal transmission through blood transfusion or sexual contact. Approximately 5% of infected individuals develop adult T-cell leukemia/lymphoma (ATL) with a poor prognosis, while 95% of infected individuals remain asymptomatic for the rest of their lives, during which time the infected cells maintain a stable immortalized latent state in the body. It is not known why such a long latent state is maintained. We hypothesize that the role of functional proteins of HTLV-1 during early infection influences the phenotype of infected cells in latency. In eukaryotic cells, a mRNA quality control mechanism called nonsense-mediated mRNA decay (NMD) functions not only to eliminate abnormal mRNAs with nonsense codons but also to target virus-derived RNAs. We have reported that HTLV-1 genomic RNA is a potential target of NMD, and that Rex suppresses NMD and stabilizes viral RNA against it. In this study, we aimed to elucidate the molecular mechanism of NMD suppression by Rex using various Rex mutant proteins. We found that region X (aa20-57) of Rex, the function of which has not been clarified, is required for NMD repression. We showed that Rex binds to Upf1, which is the host key regulator to detect abnormal mRNA and initiate NMD, through this region. Rex also interacts with SMG5 and SMG7, which play essential roles for the completion of the NMD pathway. Moreover, Rex selectively binds to Upf3B, which is involved in the normal NMD complex, and replaces it with a less active form, Upf3A, to reduce NMD activity. These results revealed that Rex invades the NMD cascade from its initiation to completion and suppresses host NMD activity to protect the viral genomic mRNA.


Asunto(s)
Productos del Gen rex/metabolismo , Virus Linfotrópico T Tipo 1 Humano/fisiología , Degradación de ARNm Mediada por Codón sin Sentido , Proteínas Portadoras/metabolismo , Línea Celular , Productos del Gen rex/genética , Genoma Viral/genética , Humanos , Carioferinas/metabolismo , Mutación , Fosforilación , Unión Proteica , Dominios Proteicos , ARN Helicasas/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/metabolismo , Proteína Exportina 1
3.
Viruses ; 14(2)2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35216000

RESUMEN

After integration to the human genome as a provirus, human T-cell leukemia virus type 1 (HTLV-1) utilizes host T cell gene expression machinery for viral replication. The viral RNA-binding protein, Rex, is known to transport unspliced/incompletely spliced viral mRNAs encoding viral structural proteins out of the nucleus to enhance virus particle formation. However, the detailed mechanism of how Rex avoids extra splicing of unspliced/incompletely spliced viral mRNAs and stabilizes them for effective translation is still unclear. To elucidate the underlying molecular mechanism of Rex function, we comprehensively analyzed the changes in gene expression and splicing patterns in Rex-overexpressing T cells. In addition, we identified 81 human proteins interacting with Rex, involved in transcription, splicing, translation, and mRNA quality control. In particular, Rex interacts with NONO and SFPQ, which play important roles in the regulation of transcription and splicing. Accordingly, expression profiles and splicing patterns of a wide variety of genes are significantly changed in Rex-expressing T cells. Especially, the level of vPD-L1 mRNA that lacks the part of exon 4, thus encodes soluble PD-L1 was significantly increased in Rex-expressing cells. Overall, by integrated analysis of these three datasets, we showed for the first time that Rex intervenes the host gene expression machinery throughout the pathway, probably to escort viral unstable mRNAs from transcription (start) to translation (end). Upon exerting its function, Rex may alter the expression level and splicing patterns of various genes, thus influencing the phenotype of the host cell.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Productos del Gen rex/metabolismo , Infecciones por HTLV-I/metabolismo , Virus Linfotrópico T Tipo 1 Humano/genética , Proteínas de Unión al ARN/metabolismo , Replicación Viral/genética , Antígeno B7-H1/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Regulación Viral de la Expresión Génica , Humanos , Factor de Empalme Asociado a PTB/metabolismo , Empalme del ARN , ARN Mensajero , Proteínas de Unión al ARN/genética
4.
Cancer Sci ; 112(6): 2542-2555, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33738869

RESUMEN

We previously indicated that Hodgkin lymphoma (HL) cells contain a small side population (SP) that differentiate into a large major population (MP) with giant Hodgkin and Reed-Sternberg (H and RS)-like cells. However, its molecular mechanisms are not fully understood. In this study, we found that intracellular reactive oxygen species (ROS) are low in the SP compared to the MP. Hydrogen peroxide induces large H- and RS-like cells in HL cell lines, but induces cell death in unrelated lymphoid cell lines. Microarray analyses revealed the enrichment of upregulated genes under hypoxic conditions in the SP compared to the MP, and we verified that the SP cells are hypoxic. Hypoxia inducible factor (HIF)-1α was preferentially expressed in the SP. CoCl2 , a HIF-1α stabilizer, blunted the effect of hydrogen peroxide. Heme oxygenase-1 (HO-1), a scavenger of ROS, was triggered by HIF-1α. The effect of hydrogen peroxide was inhibited by HO-1 induction, whereas it was promoted by HO-1 knockdown. HO-1 inhibition by zinc protoporphyrin promoted the differentiation and increased ROS. These results stress the unique roles of ROS in the differentiation of HL cells. Immature HL cells are inhibited from differentiation by a reduction of ROS through the induction of HO-1 via HIF-1α. The breakdown of this might cause the accumulation of intracellular ROS, resulting in the promotion of HL cell differentiation.


Asunto(s)
Hemo-Oxigenasa 1/genética , Enfermedad de Hodgkin/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Especies Reactivas de Oxígeno/metabolismo , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula , Línea Celular Tumoral , Cobalto/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Enfermedad de Hodgkin/genética , Humanos , Peróxido de Hidrógeno/farmacología , Protoporfirinas/farmacología
5.
Sci Rep ; 11(1): 4114, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33603066

RESUMEN

Wnt5a is a ligand of the non-canonical Wnt signaling pathway involved in cell differentiation, motility, and inflammatory response. Adult T-cell leukemia/lymphoma (ATL) is one of the most aggressive T-cell malignancies caused by infection of human T-cell leukemia virus type1 (HTLV-1). Among subtypes of ATL, acute-type ATL cells are particularly resistant to current multidrug chemotherapies and show remarkably high cell-proliferative and invasive phenotypes. Here we show a dramatic increase of WNT5A gene expression in acute-type ATL cells compared with those of indolent-type ATL cells. Treatment with IWP-2 or Wnt5a-specific knockdown significantly suppressed cell growth of ATL-derived T-cell lines. We demonstrated that the overexpression of c-Myb and FoxM1 was responsible for the synergistic activation of the WNT5A promoter. Also, a WNT5A transcript variant without the exon4 (the ΔE4-WNT5A mRNA), encoding ΔC-Wnt5 (1-136aa of 380aa), is overexpressed in acute-type ATL cells. The ΔC-Wnt5a is secreted extracellularly and enhances cellular migration/invasion to a greater extent compared with wildtype (WT)-Wnt5a. Moreover, the ΔC-Wnt5a secretion was not suppressed by IWP-2, indicating that this mutant Wnt5a is secreted via a different pathway from the WT-Wnt5a. Taken together, synergistic overexpression of the ΔC-Wnt5a by c-Myb and FoxM1 may be responsible for the malignant phenotype of acute-type ATL cells.


Asunto(s)
Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/patología , Linfocitos T/patología , Proteína Wnt-5a/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Deltaretrovirus/genética , Expresión Génica/genética , Humanos , Células Jurkat , Fenotipo , Vía de Señalización Wnt/genética
6.
Cell Rep ; 29(8): 2321-2337.e7, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31747604

RESUMEN

Although global H3K27me3 reprogramming is a hallmark of cancer, no effective therapeutic strategy for H3K27me3-high malignancies harboring EZH2WT/WT has yet been established. We explore epigenome and transcriptome in EZH2WT/WT and EZH2WT/Mu aggressive lymphomas and show that mutual interference and compensatory function of co-expressed EZH1 and EZH2 rearrange their own genome-wide distribution, thereby establishing restricted chromatin and gene expression signatures. Direct comparison of leading compounds introduces potency and a mechanism of action of the EZH1/2 dual inhibitor (valemetostat). The synthetic lethality is observed in all lymphoma models and primary adult T cell leukemia-lymphoma (ATL) cells. Opposing actions of EZH1/2-polycomb and SWI/SNF complexes are required for facultative heterochromatin formation. Inactivation of chromatin-associated genes (ARID1A, SMARCA4/BRG1, SMARCB1/SNF5, KDM6A/UTX, BAP1, KMT2D/MLL2) and oncovirus infection (HTLV-1, EBV) trigger EZH1/2 perturbation and H3K27me3 deposition. Our study provides the mechanism-based rationale for chemical dual targeting of EZH1/2 in cancer epigenome.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histonas/metabolismo , Linfoma/genética , Linfoma/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Adulto , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Epigenoma/genética , Herpesvirus Humano 4/patogenicidad , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Metilación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 2/genética , Retroviridae/patogenicidad , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
7.
Mol Cancer Res ; 17(12): 2522-2536, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31594868

RESUMEN

The genetic and molecular alterations responsible for leukemogenesis and progression of HTLV-infected adult T-cell leukemia (ATL) have not been fully clarified. Previously, we reported that various genes are not only overexpressed but also abnormally spliced in ATL cells. Here, we identified various CASP8 transcript variants in PBMCs from a smoldering-type ATL patient, which encode aberrant truncated caspase 8 (Casp8) isoforms. Among those, we focus on the three transcript variants, CASP8L (including the first 136 bp of the intron 8 between exon 8 and exon 9), CASP8-ΔE4 (without the exon 4), and CASP8-ΔE7 (without the exon 7), because they encode isoforms, Casp8L, Casp8-ΔE4, and Casp8-ΔE7, respectively, without the C-terminal catalytic domains. In this study, we conducted in vitro characterization and functional analysis of those mutant Casp8 isoforms to clarify their changed functions compared with the wild-type (WT)-Casp8. We demonstrated that these abnormal Casp8 isoforms showed lower ability to induce apoptosis than WT-Casp8 due to their dominant-negative interactions with WT-Casp8, which impair WT-Casp8 homodimerization that is essential for induction of apoptosis. Moreover, Casp8L and Casp8-ΔE7, which have only two death-effector domains, significantly activated NFκB by forming filament-like structures, which probably function as scaffolds for the IKK complex formation. In view of increasing levels of these abnormal CASP8 transcripts in primary PBMCs from HTLV-1 carriers and patients with ATL, we propose a possibility that overexpression of those Casp8 mutants, with lower proapoptotic activities and higher NFκB-activating functions than WT-Casp8, may be one of the molecular abnormalities causing malignant transformation and growth of ATL cells. IMPLICATIONS: We describe naturally occurring CASP8 transcription variants in PBMCs from patients with ATL, which encode truncated Casp8-mutant isoforms with lower proapoptotic activities and higher NFκB-activating functions compared with WT-Casp8.


Asunto(s)
Empalme Alternativo/genética , Caspasa 8/genética , Deltaretrovirus/genética , Leucemia de Células T/genética , Apoptosis/genética , Caspasa 8/sangre , Línea Celular Tumoral , Proliferación Celular/genética , Exones/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia de Células T/sangre , Leucemia de Células T/patología , Leucemia de Células T/virología , Masculino , Empalme del ARN/genética , Transducción de Señal/genética
8.
Cancer Sci ; 110(12): 3746-3753, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31642546

RESUMEN

We recently took advantage of the universal expression of cell adhesion molecule 1 (CADM1) by CD4+ cells infected with HTLV-1 and the downregulation of CD7 expression that corresponds with the oncogenic stage of HTLV-1-infected cells to develop a flow cytometric system using CADM1 versus CD7 plotting of CD4+ cells. We risk-stratified HTLV-1 asymptomatic carriers (AC) and indolent adult T-cell leukemia/lymphoma (ATL) cases based on the CADM1+ percentage, in which HTLV-1-infected clones are efficiently enriched. AC and indolent ATL cases were initially classified according to their CADM1+ cell percentage. Follow-up clinical and flow cytometric data were obtained for 71 cases. In G1 (CADM1+ ≤ 10%) and G2 (10% < CADM1+ ≤ 25%) cases, no apparent clinical disease progression was observed. In G3 (25% < CADM1+ ≤ 50%) cases, five out of nine (55.5%) cases progressed from AC to smoldering-type ATL. In G4 (50% < CADM1+ ) cases, the cumulative incidence of receiving systemic chemotherapy at 3 years was 28.4%. Our results indicate that the percentage of the CD4+ CADM1+ population predicts clinical disease progression: G1 and G2 cases, including AC cases, are stable and considered to be at low risk; G3 cases, including advanced AC cases and smoldering-type ATL cases based on the Shimoyama criteria, are considered to have intermediate risk; and G4 cases, which are mainly indolent ATL cases, are unstable and at high risk of acute transformation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Portador Sano/inmunología , Molécula 1 de Adhesión Celular/análisis , Infecciones por HTLV-I/inmunología , Leucemia-Linfoma de Células T del Adulto/inmunología , Adulto , Anciano , Progresión de la Enfermedad , Femenino , Infecciones por HTLV-I/tratamiento farmacológico , Humanos , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Masculino , Persona de Mediana Edad
9.
Anal Sci ; 34(6): 681-685, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29887556

RESUMEN

A determination of rubidium (Rb) was carried out by isotope dilution (ID) using an inductively coupled plasma tandem quadrupole mass spectrometer (ICP-QMS/QMS) with an octupole reaction-cell (ORC). Spectral interference of 87Sr with the measurement of 87Rb was effectively removed by using fluoromethane (CH3F) as the reaction cell gas at the optimum flow rate. In comparison to the measurement obtained with a mathematical correction, good precision for the analysis of the Rb isotope could be obtained independent of the concentration of Sr without any chemical separation in advance. The usefulness of the present approach was confirmed by an analysis of Rb in multiple certified reference materials (CRMs) for food and environmental analysis, for which the results agreed with their certified values in the range of expanded uncertainty.

10.
Biochem Biophys Res Commun ; 496(2): 614-620, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29330050

RESUMEN

OX40 receptor (tumor necrosis factor receptor superfamily, member 4; CD134) is a T-cell co-stimulatory molecule that plays an important role in T-cell activation and survival. OX40 receptor is activated by its ligand, OX40L; and modulation of the OX40-OX40L interaction is a promising target for the treatment of autoimmune diseases and cancers. Here, we generated a high-affinity anti-OX40 single-chain variable fragment carrying a C-terminal cysteine residue (scFvC). Physicochemical and functional analyses revealed that the scFvC bound to OX40-expressing cells and was internalized via OX40-mediated endocytosis without inducing phosphorylation of IκBα (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha), an important complex in the classical NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells) signaling pathway. In addition, mutation of the 36th cysteine residue in variable region of light chain enabled site-specific chemical modification to carboxy terminal cysteine and improved the thermal stability of the scFvC. These results suggest that this novel high-affinity anti-OX40 scFvC may be useful as a transporter for targeted delivery of small compounds, proteins, peptides, liposomes, and nanoparticles, into OX40-expressing cells for the treatment of autoimmune diseases and cancers.


Asunto(s)
Inmunoconjugados/inmunología , Receptores OX40/inmunología , Anticuerpos de Cadena Única/inmunología , Línea Celular , Sistemas de Liberación de Medicamentos , Escherichia coli/genética , Expresión Génica , Humanos , Inmunoconjugados/química , Inmunoconjugados/genética , Células Jurkat , Modelos Moleculares , Mutación Puntual , Estabilidad Proteica , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética
11.
Am J Pathol ; 187(1): 163-175, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27870927

RESUMEN

Previous studies report deregulation of multiple signaling pathways in classic Hodgkin lymphoma (cHL) cells. However, the mechanisms of how these pathways are integrated are not fully understood. Herein, we show involvement of cHL hallmark antigen CD30 in this process. CD30 facilitates phosphorylation of heat shock factor 1, activates heat shock promoter element, and induces heat shock protein (HSP) 90. CD30 repression and subsequent inhibition of HSP90 suppresses NF-κB, extracellular signal-regulated kinase, AKT, and STAT pathways in cHL cell lines. Thus, CD30-mediated induction of HSP90 appears to serve as a central hub for integration of intracellular signaling in cHL cells. We also show that CD30 induces HSP90 through phosphorylation of heat shock factor 1 via c-Jun N-terminal kinase in cHL cells. Although anaplastic large-cell lymphoma (ALCL) also is associated with CD30 overexpression, our experiments reveal that HSP90 induction in ALCL-bearing nucleophosmin-anaplastic lymphoma kinase (ALK) does not depend on CD30 but instead on ALK via c-Jun N-terminal kinase. Together, these results highlight a novel role for CD30 in mediating integration of signaling pathways of cHL cells while being replaced in this function by ALK in ALCL cells.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Enfermedad de Hodgkin/metabolismo , Antígeno Ki-1/metabolismo , Transducción de Señal , Quinasa de Linfoma Anaplásico , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Transcripción del Choque Térmico , Respuesta al Choque Térmico/genética , Enfermedad de Hodgkin/patología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Linfoma Anaplásico de Células Grandes/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Elementos de Respuesta/genética , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/metabolismo
12.
Int J Hematol ; 104(3): 330-7, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27383637

RESUMEN

Adult T-cell leukemia/lymphoma (ATLL) is a peripheral T-cell neoplasm caused by the transformation of HTLV-1-infected T cells. ATLL, especially its aggressive form, is known for its poor prognosis, even with intensive chemotherapy. ATLL cells are considered to be monoclonal; however, multiclonal proliferation or emergence of a new clone over time has been reported based on Southern blot analysis, although direct molecular evidence remains elusive. Furthermore, it is thought that clonal change may be a cause of early drug resistance in ATLL. To directly analyze potential clonal changes in ATLL during its clinical course, we used inverse PCR to detect integration sites in combination with a newly developed method using next-generation sequencing, and compared ATLL cell clonality at different time points. The results of inverse PCR indicated that the major clone was altered in three of 19 patients. Together with results from five patients, using this new method, we found direct evidence of clonal change occurring during the clinical course or in response to chemotherapy in ATLL. These results also highlight the importance of clonality analysis for understanding the mechanisms of ATLL development and drug resistance.


Asunto(s)
Células Clonales/patología , Leucemia-Linfoma de Células T del Adulto/patología , Adulto , Transformación Celular Neoplásica , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
13.
Clin Cancer Res ; 22(23): 5915-5928, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27307595

RESUMEN

PURPOSE: Adult T-cell leukemia/lymphoma (ATLL) is an aggressive human T-cell malignancy induced by human T-lymphotrophic virus-1 (HTLV-1) infection. The genetic alterations in infected cells that lead to transformation have not been completely elucidated, thus hindering the identification of effective therapeutic targets for ATL. Here, we present the first assessment of MYB proto-oncogene dysregulation in ATL and an exploration of its role in the onset of ATL. EXPERIMENTAL DESIGN: We investigated the expression patterns of MYB splicing variants in ATL. The molecular characteristics of the c-Myb-9A isoform, which was overexpressed in ATL cells, were examined using chromatin immunoprecipitation and promoter assays. We further examined the biologic impacts of abnormal c-Myb overexpression in ATL using overall c-Myb knockdown with shRNA or c-Myb-9A knockdown with morpholino oligomers. RESULTS: Both total c-Myb and c-Myb-9A, which exhibited strong transforming activity, were overexpressed in ATL cells in a leukemogenesis- and progression-dependent manner. Knockdown of either total c-Myb or c-Myb-9A induced ATL cell death. c-Myb transactivates nine genes that encode essential regulators of cell proliferation and NF-κB signaling. c-Myb-9A induced significantly stronger transactivation of all tested genes and stronger NF-κB activation compared with wild-type c-Myb. CONCLUSIONS: Our data demonstrate that c-Myb pathway overactivation caused by unbalanced c-Myb-9A overexpression is associated with disorders in cellular homeostasis and consequently, accelerated transformation, cell proliferation, and malignancy in ATL cells. These data support the notion of the c-Myb pathway as a promising new therapeutic target for ATL. Clin Cancer Res; 22(23); 5915-28. ©2016 AACR.


Asunto(s)
Regulación Leucémica de la Expresión Génica/genética , Leucemia-Linfoma de Células T del Adulto/genética , Proteínas Proto-Oncogénicas c-myb/genética , Proto-Oncogenes/genética , Transducción de Señal/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Células HEK293 , Infecciones por HTLV-I/genética , Infecciones por HTLV-I/virología , Células HeLa , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Leucemia-Linfoma de Células T del Adulto/virología , FN-kappa B/genética , Regiones Promotoras Genéticas/genética , Proto-Oncogenes Mas , Linfocitos T/virología
14.
Viruses ; 8(3): 58, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26927155

RESUMEN

Human T-cell leukemia virus type-1 (HTLV-1) Rex is a viral RNA binding protein. The most important and well-known function of Rex is stabilizing and exporting viral mRNAs from the nucleus, particularly for unspliced/partially-spliced mRNAs encoding the structural proteins essential for viral replication. Without Rex, these unspliced viral mRNAs would otherwise be completely spliced. Therefore, Rex is vital for the translation of structural proteins and the stabilization of viral genomic RNA and, thus, for viral replication. Rex schedules the period of extensive viral replication and suppression to enter latency. Although the importance of Rex in the viral life-cycle is well understood, the underlying molecular mechanism of how Rex achieves its function has not been clarified. For example, how does Rex protect unspliced/partially-spliced viral mRNAs from the host cellular splicing machinery? How does Rex protect viral mRNAs, antigenic to eukaryotic cells, from cellular mRNA surveillance mechanisms? Here we will discuss these mechanisms, which explain the function of Rex as an organizer of HTLV-1 expression based on previously and recently discovered aspects of Rex. We also focus on the potential influence of Rex on the homeostasis of the infected cell and how it can exert its function.


Asunto(s)
Regulación Viral de la Expresión Génica , Productos del Gen rex/metabolismo , Interacciones Huésped-Patógeno , Virus Linfotrópico T Tipo 1 Humano/fisiología , Replicación Viral , Humanos , Biosíntesis de Proteínas , Empalme del ARN , ARN Viral/metabolismo
15.
Blood ; 127(14): 1790-802, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26773042

RESUMEN

Adult T-cell leukemia-lymphoma (ATL) shows global gene expression alterations that confer cellular characteristics and unfavorable prognosis. However, molecular mechanisms of the sustained expression changes are largely unknown, because there is no study addressing the relationship between landscapes of the gene expression and epigenetic modifications. Here, we analyzed ATL epigenome and integrated it with transcriptome from primary ATL cells and those from corresponding normal CD4(+)T cells to decipher ATL-specific "epigenetic code" that was critical for cell identity. We found that polycomb-repressive complex 2 (PRC2)-mediated trimethylation at histone H3Lys27 (H3K27me3) was significantly and frequently reprogrammed at half of genes in ATL cells. A large proportion of the abnormal gene downregulation was detected at the early stage of disease progression and was explained by H3K27me3 accumulation. The global H3K27me3 alterations involved ATL-specific gene expression changes that included several tumor suppressors, transcription factors, epigenetic modifiers, miRNAs, and developmental genes, suggesting diverse outcomes by the PRC2-dependent hierarchical regulation. Interestingly, a key enzyme, EZH2, was sensitive to promiscuous signaling network including the NF-κB pathway and was functionally affected by human T-cell leukemia virus type I (HTLV-1) Tax. The Tax-dependent immortalized cells showed H3K27me3 reprogramming that was significantly similar to that of ATL cells. Of note, a majority of the epigenetic silencing has occurred in leukemic cells from indolent ATL and also in HTLV-1-infected T cells from asymptomatic HTLV-1 carriers. Because pharmacologic inhibition of EZH2 reversed epigenetic disruption and selectively eliminated leukemic and HTLV-1-infected cells, targeting the epigenetic elements will hold great promise in treatment and prevention of the onset of ATL and HTLV-1-related diseases.


Asunto(s)
Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Leucemia-Linfoma de Células T del Adulto/metabolismo , Proteínas de Neoplasias/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Adulto , Línea Celular Transformada , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Productos del Gen tax/genética , Productos del Gen tax/metabolismo , Histonas/genética , Histonas/metabolismo , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/patología , Masculino , Proteínas de Neoplasias/genética , Complejo Represivo Polycomb 2/genética
16.
Blood ; 127(5): 596-604, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26574607

RESUMEN

Adult T-cell leukemia/lymphoma (ATLL) is a distinct form of peripheral T-cell lymphoma with poor prognosis, which is caused by the human T-lymphotropic virus type 1 (HTLV-1). In contrast to the unequivocal importance of HTLV-1 infection in the pathogenesis of ATLL, the role of acquired mutations in HTLV-1 infected T cells has not been fully elucidated, with a handful of genes known to be recurrently mutated. In this study, we identified unique RHOA mutations in ATLL through whole genome sequencing of an index case, followed by deep sequencing of 203 ATLL samples. RHOA mutations showed distinct distribution and function from those found in other cancers. Involving 15% (30/203) of ATLL cases, RHOA mutations were widely distributed across the entire coding sequence but almost invariably located at the guanosine triphosphate (GTP)-binding pocket, with Cys16Arg being most frequently observed. Unexpectedly, depending on mutation types and positions, these RHOA mutants showed different or even opposite functional consequences in terms of GTP/guanosine diphosphate (GDP)-binding kinetics, regulation of actin fibers, and transcriptional activation. The Gly17Val mutant did not bind GTP/GDP and act as a dominant negative molecule, whereas other mutants (Cys16Arg and Ala161Pro) showed fast GTP/GDP cycling with enhanced transcriptional activation. These findings suggest that both loss- and gain-of-RHOA functions could be involved in ATLL leukemogenesis. In summary, our study not only provides a novel insight into the molecular pathogenesis of ATLL but also highlights a unique role of variegation of heterologous RHOA mutations in human cancers.


Asunto(s)
Leucemia-Linfoma de Células T del Adulto/genética , Mutación , Proteína de Unión al GTP rhoA/genética , Adulto , Secuencia de Aminoácidos , Sitios de Unión , Análisis Mutacional de ADN , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/patología , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Proteína de Unión al GTP rhoA/química , Proteína de Unión al GTP rhoA/metabolismo
17.
Sci Rep ; 5: 17868, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26639163

RESUMEN

Biological robustness is exposed to stochastic perturbations, which should be controlled by intrinsic mechanisms; the promiscuous signaling network without appropriate alleviation is the true nature of cancer cells. B cell receptor (BCR) signaling is a major source of gene expression signature important for B cell. It is still unclear the mechanism by which the expression of functionally important genes is continuously deregulated in malignant lymphomas. Using RISC-capture assay, we reveal that multiple BCR signaling factors are persistently regulated by microRNA (miRNA) in human B cells. Clinical samples from patients with diffuse large B-cell lymphoma (DLBCL, n = 83) show loss of an essential miRNA set (miR-200c, miR-203, miR-31). Conventional screening and RISC profiling identify multiple targets (CD79B, SYK, PKCßII, PLCγ1, IKKß, NIK, MYD88, PI3K class I (α/ß/δ/γ), RasGRP3); signaling network habitually faces interference composed by miRNA group in normal B cells. We demonstrate that simultaneous depletion of the key miRNAs enhances translation of the multiple targets and causes chronic activation of NF-κB, PI3K-Akt, and Ras-Erk cascades, leading to B cell transformation. This study suggests that compensatory actions by multiple miRNAs rather than by a single miRNA ensure robustness of biological processes.


Asunto(s)
MicroARNs/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/genética , Bioensayo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Linfoma de Células B Grandes Difuso/genética , MicroARNs/genética
18.
Mol Ther Methods Clin Dev ; 2: 15008, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26029719

RESUMEN

The prometastatic calcium-binding protein, S100A4, is expressed in endothelial cells, and its downregulation markedly suppresses tumor angiogenesis in a xenograft cancer model. Given that endothelial S100A4 can be a molecular target for inhibiting tumor angiogenesis, we addressed here whether synthetic peptide capable of blocking S100A4-effector protein interaction could be a novel antiangiogenic agent. To examine this hypothesis, we focused on the S100A4-binding domain of methionine aminopeptidase 2, an effector protein, which plays a role in endothelial cell growth. Overexpression of the domain in mouse endothelial MSS31 cells reduced DNA synthesis, and the corresponding synthetic peptide (named NBD) indeed interacted with S100A4 and inhibited capillary formation in vitro and new blood vessel formation in vivo. Intriguingly, a single intra-tumor administration of the NBD peptide in human prostate cancer xenografts significantly reduced vascularity, resulting in tumor regression. Mechanistically, the NBD peptide enhanced assembly of nonmuscle myosin IIA filaments along with Ser1943 phosphorylation, stimulated formation of focal adhesions without phosphorylation of focal adhesion kinase, and provoked G1/S arrest of the cell cycle. Altogether, the NBD peptide is a potent inhibitor for tumor angiogenesis, and is the first example of an anticancer peptide drug developed on the basis of an endothelial S100A4-targeted strategy.

19.
Cancer Sci ; 106(5): 598-603, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25703103

RESUMEN

We previously reported that the cell adhesion molecule 1 (CADM1) versus CD7 plot in flow cytometry reflects disease progression in human T-cell leukemia virus type 1 (HTLV-1) infection. In CD4(+) cells from peripheral blood, CADM1(-) CD7(+) (P), CADM1(+) CD7(dim) (D) and CADM1(+) CD7(-) (N) subpopulations are observed. The D and N subpopulations increase as asymptomatic HTLV-1 carriers (AC) progress to indolent adult T-cell leukemia-lymphoma (ATL) and the N subpopulation then expands in aggressive ATL. In the present study we examined whether the analysis can estimate the risk of developing ATL in advanced AC. Peripheral blood samples from AC (N = 41) and indolent ATL patients (N = 19) were analyzed by flow cytometry using the CADM1 versus CD7 plot for CD4(+) cells and inverse long PCR (clonality analysis) of FACS-sorted subpopulations. Almost all AC with a high HTLV-1 proviral load (>4 copies/100 cells) had a CADM1(+) (D + N) frequency of >10%. AC with 25% < CADM1(+) ≤ 50% contained expanded clones similar to smoldering-type ATL. In many patients in the 25% < CADM1(+) ≤ 50% group, the proportion of abnormal lymphocytes was distributed around the 5% line, which divides AC and smoldering-type ATL in Shimoyama's classification. In conclusion, the CADM1 versus CD7 plot is useful for selection of putative high-risk AC. The characteristics of some AC and smoldering ATL are said to be similar; however, long-term follow up is required and the clinical outcome (e.g. rate of transformation) of these cases should be used to determine whether to include them in the same clinical category.


Asunto(s)
Moléculas de Adhesión Celular/sangre , Citometría de Flujo/métodos , Infecciones por HTLV-I/patología , Virus Linfotrópico T Tipo 1 Humano/aislamiento & purificación , Inmunoglobulinas/sangre , Leucemia-Linfoma de Células T del Adulto/patología , Adulto , Anciano , Antígenos CD7/sangre , Molécula 1 de Adhesión Celular , Femenino , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Leucemia-Linfoma de Células T del Adulto/diagnóstico , Leucemia-Linfoma de Células T del Adulto/virología , Linfocitos/patología , Linfocitos/virología , Masculino , Persona de Mediana Edad , Carga Viral
20.
Cancer Sci ; 105(9): 1160-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24996003

RESUMEN

One of the hallmarks of cancer, global gene expression alteration, is closely associated with the development and malignant characteristics associated with adult T-cell leukemia (ATL) as well as other cancers. Here, we show that aberrant overexpression of the Ellis Van Creveld (EVC) family is responsible for cellular Hedgehog (HH) activation, which provides the pro-survival ability of ATL cells. Using microarray, quantitative RT-PCR and immunohistochemistry we have demonstrated that EVC is significantly upregulated in ATL and human T-cell leukemia virus type I (HTLV-1)-infected cells. Epigenetic marks, including histone H3 acetylation and Lys4 trimethylation, are specifically accumulated at the EVC locus in ATL samples. The HTLV-1 Tax participates in the coordination of EVC expression in an epigenetic fashion. The treatment of shRNA targeting EVC, as well as the transcription factors for HH signaling, diminishes the HH activation and leads to apoptotic death in ATL cell lines. We also showed that a HH signaling inhibitor, GANT61, induces strong apoptosis in the established ATL cell lines and patient-derived primary ATL cells. Therefore, our data indicate that HH activation is involved in the regulation of leukemic cell survival. The epigenetically deregulated EVC appears to play an important role for HH activation. The possible use of EVC as a specific cell marker and a novel drug target for HTLV-1-infected T-cells is implicated by these findings. The HH inhibitors are suggested as drug candidates for ATL therapy. Our findings also suggest chromatin rearrangement associated with active histone markers in ATL.


Asunto(s)
Epigénesis Genética , Proteínas Hedgehog/metabolismo , Leucemia de Células T/genética , Proteínas/genética , Antineoplásicos/farmacología , Secuencia de Bases , Estudios de Casos y Controles , Supervivencia Celular , Islas de CpG , Metilación de ADN , Regulación Leucémica de la Expresión Génica , Productos del Gen tax/fisiología , Células HEK293 , Humanos , Células Jurkat , Leucemia de Células T/metabolismo , Proteínas de la Membrana , Datos de Secuencia Molecular , Proteínas/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Análisis de Secuencia de ADN , Transducción de Señal , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA