Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 33(13-14): 763-781, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31123064

RESUMEN

Coordinated induction, but also repression, of genes are key to normal differentiation. Although the role of lineage-specific transcription regulators has been studied extensively, their functional integration with chromatin remodelers, one of the key enzymatic machineries that control chromatin accessibility, remains ill-defined. Here we investigate the role of Mi-2ß, a SNF-2-like nucleosome remodeler and key component of the nucleosome remodeling and histone deacetylase (NuRD) complex in early B cells. Inactivation of Mi-2ß arrested differentiation at the large pre-B-cell stage and caused derepression of cell adhesion and cell migration signaling factors by increasing chromatin access at poised enhancers and chromosome architectural elements. Mi-2ß also supported IL-7R signaling, survival, and proliferation by repressing negative effectors of this pathway. Importantly, overexpression of Bcl2, a mitochondrial prosurvival gene and target of IL-7R signaling, partly rescued the differentiation block caused by Mi-2ß loss. Mi-2ß stably associated with chromatin sites that harbor binding motifs for IKAROS and EBF1 and physically associated with these transcription factors both on and off chromatin. Notably, Mi-2ß shared loss-of-function cellular and molecular phenotypes with IKAROS and EBF1, albeit in a distinct fashion. Thus, the nucleosome remodeler Mi-2ß promotes pre-B-cell differentiation by providing repression capabilities to distinct lineage-specific transcription factor-based regulatory networks.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular/genética , Cromatina/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Linaje de la Célula , Proliferación Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Ratones , Factores de Transcripción
2.
Nat Immunol ; 15(3): 294-304, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24509510

RESUMEN

Deletion of the DNA-binding domain of the transcription factor Ikaros generates dominant-negative isoforms that interfere with its activity and correlate with poor prognosis in human precursor B cell acute lymphoblastic leukemia (B-ALL). Here we found that conditional inactivation of the Ikaros DNA-binding domain in early pre-B cells arrested their differentiation at a stage at which integrin-dependent adhesion to niches augmented signaling via mitogen-activated protein kinases, proliferation and self-renewal and attenuated signaling via the pre-B cell signaling complex (pre-BCR) and the differentiation of pre-B cells. Transplantation of polyclonal Ikaros-mutant pre-B cells resulted in long-latency oligoclonal pre-B-ALL, which demonstrates that loss of Ikaros contributes to multistep B cell leukemogenesis. Our results explain how normal pre-B cells transit from a highly proliferative and stroma-dependent phase to a stroma-independent phase during which differentiation is enabled, and suggest potential therapeutic strategies for Ikaros-mutant B-ALL.


Asunto(s)
Diferenciación Celular/inmunología , Proliferación Celular , Factor de Transcripción Ikaros/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células Precursoras de Linfocitos B/patología , Traslado Adoptivo , Animales , Apoptosis , Separación Celular , Supervivencia Celular , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factor de Transcripción Ikaros/metabolismo , Immunoblotting , Ratones , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo
3.
J Immunol ; 208(12): 2613-2621, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35623662

RESUMEN

Keratinocytes, the epithelial cells of the skin, reprogram their gene expression and produce immune effector molecules when exposed to environmental and endogenous triggers of inflammation. It remains unclear how keratinocytes process physiological signals generated during skin irritation and switch from a homeostatic to an inflammatory state. In this article, we show that the stress-activated protein kinase p38α is crucial for keratinocytes to prompt changes in their transcriptome upon cytokine stimulation and drive inflammation in allergen-exposed skin. p38α serves this function by phosphorylating p63, a transcription factor essential for the lineage identity and stemness of the skin epithelium. Phosphorylation by p38α alters the activity of p63 and redeploys this developmental transcription factor to a gene expression program linked to inflammation. Genetic ablation and pharmacological inhibition of p38α or the p38α-p63 target gene product MMP13 attenuate atopic dermatitis-like disease in mice. Our study reveals an epithelial molecular pathway promoting skin inflammation and actionable through treatment with topical small-molecule therapeutics.


Asunto(s)
Dermatitis Atópica , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Factores de Transcripción , Animales , Dermatitis Atópica/metabolismo , Inflamación/metabolismo , Queratinocitos/metabolismo , Ratones , Fosforilación , Factores de Transcripción/metabolismo
4.
Genes Dev ; 30(17): 1971-90, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27664237

RESUMEN

IKAROS is required for the differentiation of highly proliferative pre-B-cell precursors, and loss of IKAROS function indicates poor prognosis in precursor B-cell acute lymphoblastic leukemia (B-ALL). Here we show that IKAROS regulates this developmental stage by positive and negative regulation of superenhancers with distinct lineage affiliations. IKAROS defines superenhancers at pre-B-cell differentiation genes together with B-cell master regulators such as PAX5, EBF1, and IRF4 but is required for a highly permissive chromatin environment, a function that cannot be compensated for by the other transcription factors. IKAROS is also highly enriched at inactive enhancers of genes normally expressed in stem-epithelial cells. Upon IKAROS loss, expression of pre-B-cell differentiation genes is attenuated, while a group of extralineage transcription factors that are directly repressed by IKAROS and depend on EBF1 relocalization at their enhancers for expression is induced. LHX2, LMO2, and TEAD-YAP1, normally kept separate from native B-cell transcription regulators by IKAROS, now cooperate directly with them in a de novo superenhancer network with its own feed-forward transcriptional reinforcement. Induction of de novo superenhancers antagonizes Polycomb repression and superimposes aberrant stem-epithelial cell properties in a B-cell precursor. This dual mechanism of IKAROS regulation promotes differentiation while safeguarding against a hybrid stem-epithelial-B-cell phenotype that underlies high-risk B-ALL.


Asunto(s)
Diferenciación Celular/genética , Elementos de Facilitación Genéticos/fisiología , Células Epiteliales/citología , Regulación Leucémica de la Expresión Génica , Factor de Transcripción Ikaros/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/fisiopatología , Células Precursoras de Linfocitos B/citología , Animales , Epigénesis Genética , Células Epiteliales/patología , Factor de Transcripción Ikaros/genética , Ratones , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Células Precursoras de Linfocitos B/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Nat Immunol ; 13(1): 86-94, 2011 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-22080921

RESUMEN

Cell fate depends on the interplay between chromatin regulators and transcription factors. Here we show that activity of the Mi-2ß nucleosome-remodeling and histone-deacetylase (NuRD) complex was controlled by the Ikaros family of lymphoid lineage-determining proteins. Ikaros, an integral component of the NuRD complex in lymphocytes, tethered this complex to active genes encoding molecules involved in lymphoid differentiation. Loss of Ikaros DNA-binding activity caused a local increase in chromatin remodeling and histone deacetylation and suppression of lymphoid cell-specific gene expression. Without Ikaros, the NuRD complex also redistributed to transcriptionally poised genes that were not targets of Ikaros (encoding molecules involved in proliferation and metabolism), which induced their reactivation. Thus, release of NuRD from Ikaros regulation blocks lymphocyte maturation and mediates progression to a leukemic state by engaging functionally opposing epigenetic and genetic networks.


Asunto(s)
Linfocitos/enzimología , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/genética , Ensamble y Desensamble de Cromatina , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Leucemia/genética , Linfocitos/inmunología , Ratones , Motivos de Nucleótidos , Unión Proteica , Timocitos/metabolismo
6.
PLoS Genet ; 15(3): e1007765, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30897080

RESUMEN

Rare individuals with inactivating mutations in the Huntington's disease gene (HTT) exhibit variable abnormalities that imply essential HTT roles during organ development. Here we report phenotypes produced when increasingly severe hypomorphic mutations in the murine HTT orthologue Htt, (HdhneoQ20, HdhneoQ50, HdhneoQ111), were placed over a null allele (Hdhex4/5). The most severe hypomorphic allele failed to rescue null lethality at gastrulation, while the intermediate, though still severe, alleles yielded recessive perinatal lethality and a variety of fetal abnormalities affecting body size, skin, skeletal and ear formation, and transient defects in hematopoiesis. Comparative molecular analysis of wild-type and Htt-null retinoic acid-differentiated cells revealed gene network dysregulation associated with organ development that nominate polycomb repressive complexes and miRNAs as molecular mediators. Together these findings demonstrate that Htt is required both pre- and post-gastrulation to support normal development.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Alelos , Animales , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Frecuencia de los Genes/genética , Genotipo , Proteína Huntingtina/fisiología , Ratones/embriología , Mutación , Proteínas del Tejido Nervioso/genética , Fenotipo
8.
Immunity ; 30(4): 493-507, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19345118

RESUMEN

The mechanisms regulating lineage potential during early hematopoiesis were investigated. First, a cascade of lineage-affiliated gene expression signatures, primed in hematopoietic stem cells (HSCs) and differentially propagated in lineage-restricted progenitors, was identified. Lymphoid transcripts were primed as early as the HSC, together with myeloid and erythroid transcripts. Although this multilineage priming was resolved upon subsequent lineage restrictions, an unexpected cosegregation of lymphoid and myeloid gene expression and potential past a nominal myeloid restriction point was identified. Finally, we demonstrated that whereas the zinc finger DNA-binding factor Ikaros was required for induction of lymphoid lineage priming in the HSC, it was also necessary for repression of genetic programs compatible with self-renewal and multipotency downstream of the HSC. Taken together, our studies provide new insight into the priming and restriction of lineage potentials during early hematopoiesis and identify Ikaros as a key bivalent regulator of this process.


Asunto(s)
Linaje de la Célula , Redes Reguladoras de Genes , Genoma , Células Madre Hematopoyéticas/inmunología , Factor de Transcripción Ikaros/metabolismo , Linfocitos/inmunología , Animales , Regulación de la Expresión Génica , Hematopoyesis/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
9.
Curr Opin Hematol ; 24(4): 345-352, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28463873

RESUMEN

PURPOSE OF REVIEW: Loss of IKAROS in committed B cell precursors causes a block in differentiation while at the same time augments aberrant cellular properties, such as bone marrow stromal adhesion, self-renewal and resistance to glucocorticoid-mediated cell death. B cell acute lymphoblastic leukaemias originating from these early stages of B cell differentiation and associated with IKAROS mutations share a high-risk cellular phenotype suggesting that deregulation of IKAROS-based mechanisms cause a highly malignant disease process. RECENT STUDIES: Recent studies show that IKAROS is critical for the activity of super-enhancers at genes required for pre-B cell receptor (BCR) signalling and differentiation, working either downstream of or in parallel with B cell master regulators such as EBF1 and PAX5. IKAROS also directly represses a cryptic regulatory network of transcription factors prevalent in mesenchymal and epithelial precursors that includes YAP1, TEAD1/2, LHX2 and LMO2, and their targets, which are not normally expressed in lymphocytes. IKAROS prevents not only expression of these 'extra-lineage' transcription factors but also their cooperation with endogenous B cell master regulators, such as EBF1 and PAX5, leading to the formation of a de novo for lymphocytes super-enhancer network. IKAROS coordinates with the Polycomb repression complex (PRC2) to provide stable repression of associated genes during B cell development. However, induction of regulatory factors normally repressed by IKAROS starts a feed-forward loop that activates de-novo enhancers and elevates them to super-enhancer status, thereby diminishing PRC2 repression and awakening aberrant epithelial-like cell properties in B cell precursors. SUMMARY: Insight into IKAROS-based transcriptional circuits not only sets new paradigms for cell differentiation but also provides new approaches for classifying and treating high-risk human B-ALL that originates from these early stages of B cell differentiation.


Asunto(s)
Linfocitos B/metabolismo , Transformación Celular Neoplásica/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Transcripción Genética , Animales , Linfocitos B/patología , Diferenciación Celular/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Elementos de Facilitación Genéticos , Humanos , Factor de Transcripción Ikaros/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Unión Proteica
10.
Blood ; 122(18): 3149-59, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24002445

RESUMEN

Ikaros is a critical regulator of lymphocyte development and homeostasis; thus, understanding its transcriptional regulation is important from both developmental and clinical perspectives. Using a mouse transgenic reporter approach, we functionally characterized a network of highly conserved cis-acting elements at the Ikzf1 locus. We attribute B-cell and myeloid but not T-cell specificity to the main Ikzf1 promoter. Although this promoter was unable to counter local chromatin silencing effects, each of the 6 highly conserved Ikzf1 intronic enhancers alleviated silencing. Working together, the Ikzf1 enhancers provided locus control region activity, allowing reporter expression in a position and copy-independent manner. Only 1 of the Ikzf1 enhancers was responsible for the progressive upregulation of Ikaros expression from hematopoietic stem cells to lymphoid-primed multipotent progenitors to T-cell precursors, which are stages of differentiation dependent on Ikaros for normal outcome. Thus, Ikzf1 is regulated by both epigenetic and transcriptional factors that target its enhancers in both redundant and specific fashions to provide an expression profile supportive of normal lymphoid lineage progression and homeostasis. Mutations in the Ikzf1 regulatory elements and their interacting factors are likely to have adverse effects on lymphopoiesis and contribute to leukemogenesis.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Factor de Transcripción Ikaros/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Activación Transcripcional , Animales , Linfocitos B/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Encéfalo/metabolismo , Epigénesis Genética , Citometría de Flujo , Redes Reguladoras de Genes , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factor de Transcripción Ikaros/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Datos de Secuencia Molecular , Células Mieloides/metabolismo , Homología de Secuencia de Aminoácido , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo
11.
Gan To Kagaku Ryoho ; 40(12): 2470-2, 2013 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-24394147

RESUMEN

A 67-year-old man with bladder cancer who was treated with transurethral resection of bladder tumour(TUR-Bt)and chemotherapy at the age of 59 years was diagnosed as having urothelial cancer by biopsy 8 years later. Detailed examination revealed the presence of synchronous triple cancer, with hepatocellular cancer and gastric cancer. Subsequently, semi-total gastrectomy, partial hepatectomy(S6), radio frequency ablation(S5, S7), and cholecystectomy were performed. Histologically, the gastric tumor was a moderately differentiated tubular adenocarcinoma, the hepatic tumor was a moderately differentiated hepatocellular carcinoma, the bladder tumor was a transitional cell carcinoma, and the ureteral tumor was an urothelial carcinoma.


Asunto(s)
Neoplasias Hepáticas/patología , Neoplasias Primarias Múltiples/patología , Neoplasias Gástricas/patología , Neoplasias de la Vejiga Urinaria/patología , Neoplasias Urológicas/patología , Anciano , Antimetabolitos Antineoplásicos/uso terapéutico , Quimioterapia Adyuvante , Combinación de Medicamentos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/cirugía , Masculino , Neoplasias Primarias Múltiples/tratamiento farmacológico , Neoplasias Primarias Múltiples/cirugía , Ácido Oxónico/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/cirugía , Tegafur/uso terapéutico , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/cirugía , Neoplasias Urológicas/tratamiento farmacológico , Neoplasias Urológicas/cirugía
12.
Med Mol Morphol ; 45(3): 173-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23001300

RESUMEN

Extra-gastrointestinal stromal tumors (E-GISTs) not associated with the alimentary tract in the pelvic cavity are extremely rare. We treated a 49-year-old Japanese man with such an E-GIST in the pelvic cavity who underwent an intrapelvic tumorectomy with a total prostatectomy and partial rectum resection. Gross examination of the specimen revealed an 8.1 × 5 × 4 cm white-grayish mass. Histological findings showed uniform spindle cells with scant atypia that formed interlacing bundles or whorl patterns. These neoplastic cells did not invade adjacent organs, including the gut. Immunohistochemical findings revealed that the neoplastic cells were positive for c-kit, CD34, and vimentin. Molecular analysis showed a c-kit mutation at exon 9 with duplication of Ala and Tyr. Our diagnosis was E-GIST, which belongs to the intermediate group of GIST. Following the operation, we administered imatinib mesylate for 6 months. After stopping for 5 months, it was administered again for local recurrence. We are planning our future strategy for this case including surgical resection as necessary.


Asunto(s)
Pelvis , Antígenos CD34/genética , Antígenos CD34/metabolismo , Antineoplásicos/uso terapéutico , Benzamidas , Exones/genética , Tumores del Estroma Gastrointestinal/diagnóstico , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/patología , Tumores del Estroma Gastrointestinal/cirugía , Humanos , Mesilato de Imatinib , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mutación , Pelvis/patología , Pelvis/cirugía , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/uso terapéutico , Vimentina/genética , Vimentina/metabolismo
13.
Nat Commun ; 13(1): 1859, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35388001

RESUMEN

The cohesin complex participates in the organization of 3D genome through generating and maintaining DNA loops. Stromal antigen 2 (STAG2), a core subunit of the cohesin complex, is frequently mutated in various cancers. However, the impact of STAG2 inactivation on 3D genome organization, especially the long-range enhancer-promoter contacts and subsequent gene expression control in cancer, remains poorly understood. Here we show that depletion of STAG2 in melanoma cells leads to expansion of topologically associating domains (TADs) and enhances the formation of acetylated histone H3 lysine 27 (H3K27ac)-associated DNA loops at sites where binding of STAG2 is switched to its paralog STAG1. We further identify Interferon Regulatory Factor 9 (IRF9) as a major direct target of STAG2 in melanoma cells via integrated RNA-seq, STAG2 ChIP-seq and H3K27ac HiChIP analyses. We demonstrate that loss of STAG2 activates IRF9 through modulating the 3D genome organization, which in turn enhances type I interferon signaling and increases the expression of PD-L1. Our findings not only establish a previously unknown role of the STAG2 to STAG1 switch in 3D genome organization, but also reveal a functional link between STAG2 and interferon signaling in cancer cells, which may enhance the immune evasion potential in STAG2-mutant cancer.


Asunto(s)
Proteínas Cromosómicas no Histona , Melanoma , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Genoma , Humanos , Interferones/genética , Melanoma/genética
14.
Mol Cancer Res ; 19(3): 507-515, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33234577

RESUMEN

Hematopoietic-derived cells are integral components of the tumor microenvironment and serve as critical mediators of tumor-host interactions. Host cells derived from myeloid and lymphoid lineages perform well-established functions linked to cancer development, progression, and response to therapy. It is unclear whether host erythroid cells also contribute to shaping the path that cancer can take, but emerging evidence points to this possibility. Here, we show that tumor-promoting environmental stress and tumor-induced hemodynamic changes trigger renal erythropoietin production and erythropoietin-dependent expansion of splenic erythroid cell populations in mice. These erythroid cells display molecular features indicative of an immature erythroid phenotype, such as the expression of both CD71 and TER119 and the retention of intact nuclei, and express genes encoding immune checkpoint molecules. Nucleated erythroid cells with similar properties are present in mouse and human tumor tissues. Antibody-mediated erythropoietin blockade reduces tumor-responsive erythroid cell induction and tumor growth. These findings reveal the potential of tumor-induced erythropoietin and erythroid cells as targets for cancer treatment. IMPLICATIONS: : Our study identifies erythropoietin and erythroid cells as novel players in tumor-host interactions and highlights the involvement of multiorgan signaling events in their induction in response to environmental stress and tumor growth.


Asunto(s)
Células Eritroides/metabolismo , Proteínas de Punto de Control Inmunitario/metabolismo , Animales , Diferenciación Celular , Humanos , Ratones , Transducción de Señal
15.
Curr Opin Immunol ; 19(2): 116-22, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17307348

RESUMEN

Hematopoiesis is the developmental process by which all blood and immune cells are generated. A decade-old scheme has supported an early and complete separation of the erythro-myeloid from the lymphoid lineages. Recent advances have re-drawn this map, separating lymphoid and myeloid from erythroid programs early in development. Subsequently, the fate restriction of both the lympho-myeloid and the erythro-megakaryocyte progenitors is dependent on Ikaros and its associated chromatin regulators. Genetic studies of this family of nuclear factors are now providing unique insight into the functional molecular signatures that bestow plasticity to the hematopoietic stem cell and its early progeny.


Asunto(s)
Cromatina/genética , Células Precursoras Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Factor de Transcripción Ikaros/fisiología , Células Madre Multipotentes/metabolismo , Animales , Factor de Transcripción Ikaros/genética , Ratones
16.
Intern Med ; 57(19): 2807-2812, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-29780135

RESUMEN

Objective There are few reports on the outcomes of 12-week paritaprevir, ombitasvir, and ritonavir (PTV/OBV/r) treatment in real-world clinical settings. We aimed to evaluate the efficacy and safety of 12-week treatment with ritonavir-boosted paritaprevir and ombitasvir in patients with hepatitis C virus (HCV) genotype 1 infection in a real-world setting. Methods Fifty-eight patients with chronic hepatitis or compensated hepatic cirrhosis and genotype-1 HCV infection were treated with PTV/OBV/r and followed for 24 weeks after the completion of treatment in 10 centers in northern Tohoku. The efficacy and safety of this 12-week treatment regimen was analyzed. Results Among the 58 treated patients, 18 (31%) had compensated liver cirrhosis, while 11 (19%) patients had experienced treatment failure with another treatment regimen. NS5A resistance-associated variants (RAVs) were detected at baseline in 3 patients (5.2%), including Y93H in two patients and L31M in two patients. One patient had NS5A RAVs at both positions 93 and 31. The overall sustained virological response (SVR) 24 rate was 96.6%. Three patients with NS5A RAVs also achieved an SVR24. The SVR24 rate was not significantly affected by age, sex, prior treatment, prior history of HCC, or liver stiffness. The mean alanine aminotransferase (ALT) levels decreased significantly during this treatment. Adverse events occurred in 15 patients (26%), 26% of which were grade 1 or 2. No severe adverse events occurred. Conclusion In this real-world study, 12-week PTV/OBV/r treatment was effective and safe for treating patients with HCV-1 infection who had chronic hepatitis or compensated hepatic cirrhosis.


Asunto(s)
Anilidas/uso terapéutico , Antivirales/uso terapéutico , Carbamatos/uso terapéutico , Hepatitis C Crónica/tratamiento farmacológico , Compuestos Macrocíclicos/uso terapéutico , Ritonavir/uso terapéutico , Adulto , Anciano , Alanina Transaminasa , Estudios de Cohortes , Ciclopropanos , Quimioterapia Combinada , Femenino , Genotipo , Hepacivirus/genética , Humanos , Lactamas Macrocíclicas , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/virología , Masculino , Persona de Mediana Edad , Prolina/análogos & derivados , Sulfonamidas , Resultado del Tratamiento , Valina
17.
PLoS One ; 10(7): e0131568, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26135129

RESUMEN

Ikaros DNA binding factor plays critical roles in lymphocyte development. Changes in Ikaros expression levels during lymphopoiesis are controlled by redundant but also unique regulatory elements of its locus that are critical for this developmental process. We have recently shown that Ikaros binds its own locus in thymocytes in vivo. Here, we evaluated the role of an Ikaros binding site within its major lympho-myeloid promoter. We identified an Ikaros/Ets binding site within a promoter sub-region that was highly conserved in mouse and human. Deletion of this binding site increased the percentage of the reporter-expressing mouse lines, indicating that its loss provided a more permissive chromatin environment. However, once transcription was established, the lack of this site decreased transcriptional activity. These findings implicate a dual role for Ikaros/Ets1 binding on Ikzf1 expression that is exerted at least through its promoter.


Asunto(s)
Epigénesis Genética , Factor de Transcripción Ikaros/genética , Regiones Promotoras Genéticas , Transcripción Genética , Secuencias de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Inmunoprecipitación de Cromatina , Femenino , Citometría de Flujo , Eliminación de Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Factor de Transcripción Ikaros/metabolismo , Linfocitos/citología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Unión Proteica , Timocitos/metabolismo
18.
J Med Ultrason (2001) ; 29(2): 71-7, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27277743

RESUMEN

A53-year-old woman was hospitalized with a hepatic tumor that had been detected on health-screening ultrasonography. Laboratory data showed no marked findings: HBs-Ag and HCV-Ab were negative, and levels of serum AFP, PIVKA-II, CEA, and CA 19-0 were within their normal ranges. Ultrasonogram showed a hyperechoic circle concentric with and inside of a hypoechoic mass in the right hepatic lobe. The tumor appeared isodense with the surrounding liver in unenhanced CT, but enhanced CT showed a low density mass. A celiac arteriogram revealed a hypervascular tumor. The resected tumor measured 20×15 mm. Microscopic findings showed a carcinoid tumor that was positive for Grimelius stain. Further examination of the lung, gastrointestinal tract, and other organs found no primary carcinoid tumor. The patient has been well and free from tumor of the liver or elsewhere for the 7 years since her operation. We therefore considered this lesion to have been a primary carcinoid tumor of the liver. Only 14 cases of this disease have ever been reported in Japan. This report includes a review of the literature on the imaging of this rare tumor.

19.
Int J Hematol ; 100(3): 220-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25085254

RESUMEN

The Ikaros family of DNA-binding proteins are critical regulators of lymphocyte differentiation. In multipotent, hematopoietic progenitors, Ikaros supports transcriptional priming of genes promoting lymphocyte differentiation. Ikaros targets the Nucleosome Remodeling Deacetylase (NuRD) complex to lymphoid lineage genes, thereby increasing chromatin accessibility and transcriptional priming. After lymphoid lineage specification, Ikaros expression is raised to levels characteristic of intermediate B cell and T cell precursors, which is necessary to support maturation and prevent leukemogenesis. Loss of Ikaros in T cell precursors allows the NuRD complex to repress lymphocyte genes and extends its targeting to genes that support growth and proliferation, causing their activation and triggering a cascade of events that leads to leukemogenesis. Loss of Ikaros in B cell precursors blocks differentiation and perpetuates stromal adhesion by enhancing integrin signaling. The combination of integrin and cytokine signaling in Ikaros-deficient pre-B cells promotes their survival and self-renewal. The stages of lymphocyte differentiation that are highly dependent on Ikaros are underscored by changes in Ikaros transcription, supported by a complex network of stage-specific regulatory networks that converge upon the Ikzf1 locus. It is increasingly apparent that understanding the regulatory networks that operate upstream and downstream of Ikaros is critical not only for our understanding of normal lymphopoiesis, but also in placing the right finger on the mechanisms that support hematopoietic malignancies in mouse and human.


Asunto(s)
Epigénesis Genética/inmunología , Factor de Transcripción Ikaros/genética , Linfopoyesis/genética , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos T/inmunología , Animales , Autoantígenos/genética , Autoantígenos/inmunología , Diferenciación Celular , Linaje de la Célula/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Factor de Transcripción Ikaros/inmunología , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/inmunología , Ratones , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos T/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Transducción de Señal
20.
Anticancer Res ; 34(3): 1251-3, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24596368

RESUMEN

BACKGROUND/AIM: Survivin is expressed in the nucleus and/or cytoplasm of various types of malignant tumor cells. Nuclear survivin is indispensable for complete mitosis, while cytoplasmic survivin functions as an apoptosis inhibitor. We examined the difference in the survivin expression among stromal cells of fibroadenoma, and benign and malignant phyllodes tumors. MATERIALS AND METHODS: Tumor sections were immunohistochemically stained with an anti-human survivin antibody and the labeling index of survivin was calculated. RESULTS: In stromal cells of all tumors, survivin was expressed in the nuclei but not in the cytoplasm. The labeling indices of the stromal cells in five malignant phyllodes tumors (20.5±3.0) were significantly greater than those observed in eight fibroadenomas (1.9±0.6) or nine benign phyllodes tumors (3.0±0.9). CONCLUSION: In the present study it was shown that stromal cells in malignant phyllodes tumors express nuclear survivin more extensively than stromal cells in benign phyllodes tumors or fibroadenomas.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Núcleo Celular/metabolismo , Fibroadenoma/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Tumor Filoide/metabolismo , Células del Estroma/metabolismo , Neoplasias de la Mama/patología , Citoplasma/metabolismo , Femenino , Fibroadenoma/patología , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Estadificación de Neoplasias , Tumor Filoide/patología , Pronóstico , Células del Estroma/patología , Survivin
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA