Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Leukemia ; 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734786

RESUMO

TIF1ß/KAP1/TRIM28, a chromatin modulator, both represses and activates the transcription of genes in normal and malignant cells. Analyses of datasets on leukemia patients revealed that the expression level of TIF1ß was increased in patients with chronic myeloid leukemia at the blast crisis and acute myeloid leukemia. We generated a BCR::ABL1 conditional knock-in (KI) mouse model, which developed aggressive myeloid leukemia, and demonstrated that the deletion of the Tif1ß gene inhibited the progression of myeloid leukemia and showed longer survival than that in BCR::ABL1 KI mice, suggesting that Tif1ß drove the progression of BCR::ABL1-induced leukemia. In addition, the deletion of Tif1ß sensitized BCR::ABL1 KI leukemic cells to dasatinib. The deletion of Tif1ß decreased the expression levels of TIF1ß-target genes and chromatin accessibility peaks enriched with the Fosl1-binding motif in BCR::ABL1 KI stem cells. TIF1ß directly bound to the promoters of proliferation genes, such as FOSL1, in human BCR::ABL1 cells, in which TIF1ß and FOSL1 bound to adjacent regions of chromatin. Since the expression of Fosl1 was critical for the enhanced growth of BCR::ABL1 KI cells, Tif1ß and Fosl1 interacted to activate the leukemic transcriptional program in and cellular function of BCR::ABL1 KI stem cells and drove the progression of myeloid leukemia.

2.
BMC Nephrol ; 24(1): 323, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37907886

RESUMO

BACKGROUND: BK polyomavirus-associated nephropathy (BKPyVAN) has become a major cause of kidney dysfunction and graft loss in kidney transplant recipients. On rare occasion, polyomavirus has also been known to affect native kidneys of immunocompromised individuals. Only a small number of opportunistic infections have been reported in the carrier phase of human T-lymphotropic virus type 1 (HTLV-1). This is the first reported case of BKPyVAN in native kidneys of an HTLV-1 carrier. CASE PRESENTATION: A 61-year-old man was referred to our hospital from a primary care physician for work-up and treatment of pneumonia. He was diagnosed with Pneumocystis pneumonia and identified as a HTLV-1 carrier who had not yet developed adult T-cell leukemia (ATL). The pneumonia was successfully treated with sulfamethoxazole-trimethoprim. He had never been diagnosed with any kind of kidney dysfunction. Laboratory investigations showed a serum creatinine of 5.3 mg/dL, and urinary sediment showed cells with nuclear enlargement and inclusion bodies suggesting viral infection. The urinary Papanicolaou stain showed inclusions in swollen, ground-glass nuclei, typical of "decoy cells". Renal biopsy showed degeneration of tubules with epithelial enlargement, vacuolar degeneration, nuclear inclusion bodies, and detachment from the tubular basement membrane. Tubular nuclei showed positive staining positive for simian virus 40 large-T antigen. Polymerase chain reaction tests for BK polyomavirus DNA of both urine and plasma were positive. These findings confirmed a diagnosis of BKPyVAN. Intravenous immunoglobulin therapy did not improve renal function, necessitating maintenance hemodialysis therapy. CONCLUSIONS: BKPyVAN should be considered when acute kidney injury occurs with opportunistic infection. HTLV-1 carriers can develop opportunistic infections even before the onset of ATL.


Assuntos
Injúria Renal Aguda , Vírus BK , Vírus Linfotrópico T Tipo 1 Humano , Nefropatias , Transplante de Rim , Nefrite Intersticial , Infecções Oportunistas , Pneumonia , Infecções por Polyomavirus , Humanos , Masculino , Pessoa de Meia-Idade , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/complicações , Rim/patologia , Nefropatias/patologia , Transplante de Rim/efeitos adversos , Nefrite Intersticial/patologia , Infecções Oportunistas/complicações , Infecções por Polyomavirus/complicações , Infecções por Polyomavirus/diagnóstico
3.
J Exp Med ; 220(7)2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37071125

RESUMO

Aberrant innate immune signaling in myelodysplastic syndrome (MDS) hematopoietic stem/progenitor cells (HSPCs) has been implicated as a driver of the development of MDS. We herein demonstrated that a prior stimulation with bacterial and viral products followed by loss of the Tet2 gene facilitated the development of MDS via up-regulating the target genes of the Elf1 transcription factor and remodeling the epigenome in hematopoietic stem cells (HSCs) in a manner that was dependent on Polo-like kinases (Plk) downstream of Tlr3/4-Trif signaling but did not increase genomic mutations. The pharmacological inhibition of Plk function or the knockdown of Elf1 expression was sufficient to prevent the epigenetic remodeling in HSCs and diminish the enhanced clonogenicity and the impaired erythropoiesis. Moreover, this Elf1-target signature was significantly enriched in MDS HSPCs in humans. Therefore, prior infection stress and the acquisition of a driver mutation remodeled the transcriptional and epigenetic landscapes and cellular functions in HSCs via the Trif-Plk-Elf1 axis, which promoted the development of MDS.


Assuntos
Dioxigenases , Síndromes Mielodisplásicas , Humanos , Células-Tronco Hematopoéticas/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/genética , Dioxigenases/metabolismo
4.
Commun Biol ; 5(1): 1309, 2022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36446869

RESUMO

Adult T-cell leukemia/lymphoma (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1). In addition to HTLV-1 bZIP factor (HBZ), a leukemogenic antisense transcript of HTLV-1, abnormalities of genes involved in TCR-NF-κB signaling, such as CARD11, are detected in about 90% of patients. Utilizing mice expressing CD4+ T cell-specific CARD11(E626K) and/or CD4+ T cell-specific HBZ, namely CARD11(E626K)CD4-Cre mice, HBZ transgenic (Tg) mice, and CARD11(E626K)CD4-Cre;HBZ Tg double transgenic mice, we clarify these genes' pathogenetic effects. CARD11(E626K)CD4-Cre and HBZ Tg mice exhibit lymphocytic invasion to many organs, including the lungs, and double transgenic mice develop lymphoproliferative disease and increase CD4+ T cells in vivo. CARD11(E626K) and HBZ cooperatively activate the non-canonical NF-κB pathway, IRF4 targets, BATF3/IRF4/HBZ transcriptional network, MYC targets, and E2F targets. Most KEGG and HALLMARK gene sets enriched in acute-type ATL are also enriched in double transgenic mice, indicating that these genes cooperatively contribute to ATL development.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica , Leucemia-Linfoma de Células T do Adulto , Linfoma , Adulto , Animais , Humanos , Camundongos , Fatores de Transcrição de Zíper de Leucina Básica/genética , Proteínas Adaptadoras de Sinalização CARD , Guanilato Ciclase , Leucemia-Linfoma de Células T do Adulto/genética , Camundongos Transgênicos , Mutação , NF-kappa B/genética , Proteínas dos Retroviridae
5.
Biochem Biophys Res Commun ; 619: 117-123, 2022 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-35753219

RESUMO

Radiation therapy is one of the major treatment modalities for patients with cancers. However, ionizing radiation (IR) damages not only cancer cells but also the surrounding vascular endothelial cells (ECs). Hippo pathway effector genes Yap1 and Taz are the two transcriptional coactivators that have crucial roles in tissue homeostasis and vascular integrity in various organs. However, their function in adult ECs at the steady state and after IR is poorly understood. Here, we report sex- and context-dependent roles of endothelial YAP1/TAZ in maintaining vascular integrity and organismal survival. EC-specific Yap1/Taz deletion compromised systemic vascular integrity, resulting in lethal circulation failure preferentially in male mice. Furthermore, EC-specific Yap1/Taz deletion induced acute lethality upon sublethal IR that was closely associated with exacerbated systemic vascular dysfunction and circulation failure. Consistent with these findings, RNA-seq analysis revealed downregulation of tight junction genes in Yap1/Taz-deleted ECs. Collectively, our findings highlight the importance of endothelial YAP1/TAZ for maintaining adult vascular function, which may provide clinical implications for preventing organ injury after radiation therapy.


Assuntos
Neoplasias , Transativadores , Animais , Células Endoteliais/metabolismo , Masculino , Camundongos , Neoplasias/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
6.
FASEB J ; 36(7): e22345, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35635715

RESUMO

High mobility group nucleosome-binding protein 3 (HMGN3), a member of the HMGN family, modulates the structure of chromatin and regulates transcription through transcription factors. HMGN3 has been implicated in the development of various cancers; however, the underlying mechanisms remain unclear. We herein demonstrated that the high expression of HMGN3 correlated with the metastasis of liver fluke infection-induced cholangiocarcinoma (CCA) in patients in northeastern Thailand. The knockdown of HMGN3 in CCA cells significantly impaired the oncogenic properties of colony formation, migration, and invasion. HMGN3 inhibited the expression of and blocked the intracellular polarities of epithelial regulator genes, such as the CDH1/E-cadherin and TJAP1 genes in CCA cells. A chromatin immunoprecipitation sequencing analysis revealed that HMGN3 required the transcription factor SNAI2 to bind to and repress the expression of epithelial regulator genes, at least in part, due to histone deacetylases (HDACs), the pharmacological inhibition of which reactivated these epithelial regulators in CCA, leading to impairing the cell migration capacity. Therefore, the overexpression of HMGN3 represses the transcription of and blocks the polarities of epithelial regulators in CCA cells in a manner that is dependent on the SNAI2 gene and HDACs.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Regulação da Expressão Gênica , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Humanos , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Int J Hematol ; 115(4): 553-562, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35067851

RESUMO

High mobility group AT-hook 2 (Hmga2) is a chromatin modifier protein that plays a critical role in fetal development and leukemia propagation by binding to chromatin and DNA via its AT-hook domains. However, the molecular mechanisms by which Hmga2 activates the expression of target genes to drive the self-renewal of hematopoietic stem cells (HSCs) remain unclear. We generated Rosa26 locus Hmga2 conditional knock-in mice and found that overexpression of Hmga2 promoted self-renewal of normal HSCs, but maintained their fitness in bone marrow, and consequently was not sufficient to initiate malignancy. This result is consistent with previous findings showing that Hmga2 is a proto-oncogene. We also assessed the cellular functions of Hmga2 mutants lacking functional domains and demonstrated that the C-terminus acidic domain of Hmga2 and the domain's linker region were critical for activating genes involved in stem cell signatures, such as the Igf2bp2 gene, to drive proliferation of HSCs. In contrast, overexpression of Hmga1, a member of the Hmga family with a different linker region, did not drive proliferation of HSCs. Our results reveal a critical role for the acidic domain of Hmga2 and the domain's linker region in modulating the transcription and self-renewal functions of HSCs.


Assuntos
Células-Tronco Hematopoéticas , Neoplasias , Animais , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Proteínas de Ligação a RNA
8.
Cell Rep ; 34(8): 108779, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33626356

RESUMO

In the tumor microenvironment, senescent non-malignant cells, including cancer-associated fibroblasts (CAFs), exhibit a secretory profile under stress conditions; this senescence-associated secretory phenotype (SASP) leads to cancer progression and chemoresistance. However, the role of senescent CAFs in metastatic lesions and the molecular mechanism of inflammation-related SASP induction are not well understood. We show that pro-inflammatory cytokine-driven EZH2 downregulation maintains the SASP by demethylating H3K27me3 marks in CAFs and enhances peritoneal tumor formation of gastric cancer (GC) through JAK/STAT3 signaling in a mouse model. A JAK/STAT3 inhibitor blocks the increase in GC cell viability induced by senescent CAFs and peritoneal tumor formation. Single-cell mass cytometry revealed that fibroblasts exist in the ascites of GC patients with peritoneal dissemination, and the fibroblast population shows p16 expression and SASP factors at high levels. These findings provide insights into the inflammation-related SASP maintenance by histone modification and the role of senescent CAFs in GC peritoneal dissemination.


Assuntos
Fibroblastos Associados a Câncer/enzimologia , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Neoplasias Peritoneais/metabolismo , Fenótipo Secretor Associado à Senescência , Neoplasias Gástricas/metabolismo , Idoso , Animais , Antineoplásicos/farmacologia , Fibroblastos Associados a Câncer/patologia , Linhagem Celular Tumoral , Citocinas/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Inibidores de Janus Quinases/farmacologia , Janus Quinases/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Peritoneais/tratamento farmacológico , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/secundário , Piridinas/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Microambiente Tumoral , Tirfostinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Exp Hematol ; 97: 14-20, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33600870

RESUMO

RUNX3, a transcription factor, has been implicated as a tumor suppressor in various cancers, including hematological malignancies; however, recent studies revealed an oncogenic function of RUNX3 in the pathogenesis of myeloid malignancies, such as myelodysplastic syndrome and acute myeloid leukemia. In contrast to the high frequency of mutations in the RUNX1 gene, deletion of and loss-of-function mutations in RUNX3 are rarely detected in patients with hematopoietic malignancies. Although RUNX3 is expressed in normal hematopoietic stem and progenitor cells, its expression decreases with aging in humans. The loss of Runx3 did not result in the development of lethal hematological diseases in mice despite the expansion of myeloid cells. Therefore, RUNX3 does not appear to initiate the transformation of normal hematopoietic stem cells. However, the overexpression of RUNX3 inhibits the expression and transcriptional function of the RUNX1 gene, but activates the expression of key oncogenic pathways, such as MYC, resulting in the transformation of premalignant stem cells harboring a driver genetic mutation. We herein discuss the mechanisms by which RUNX3 is activated and how RUNX3 exerts oncogenic effects on the cellular function of and transcriptional program in premalignant stem cells to drive myeloid transformation.


Assuntos
Transformação Celular Neoplásica , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Leucemia Mieloide/genética , Síndromes Mielodisplásicas/genética , Células Mieloides/patologia , Animais , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Regulação Neoplásica da Expressão Gênica , Hematopoese , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/patologia , Células Mieloides/metabolismo
10.
Oncogene ; 40(8): 1531-1541, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33452460

RESUMO

High Mobility Group AT-hook 2 (HMGA2) is a chromatin modifier and its overexpression has been found in patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Level of Hmga2 expression is fine-tuned by Lin28b-Let-7 axis and Polycomb Repressive Complex 2, in which deletion of Ezh2 leads to activation of Hmga2 expression in hematopoietic stem cells. To elucidate the mechanisms by which the overexpression of HMGA2 helps transformation of stem cells harboring a driver mutation of TET2, we generated an Hmga2-expressing Tet2-deficient mouse model showing the progressive phenotypes of MDS and AML. The overexpression of Hmga2 remodeled the transcriptional program of Tet2-deficient stem and progenitor cells, leading to the impaired differentiation of myeloid cells. Furthermore, Hmga2 was bound to a proximal region of Igf2bp2 oncogene, and activated its transcription, leading to enhancing self-renewal of Tet2-deficient stem cells that was suppressed by inhibition of the DNA binding of Hmga2. These combinatory effects on the transcriptional program and cellular function were not redundant to those in Tet2-deficient cells. The present results elucidate that Hmga2 targets key oncogenic pathways during the transformation and highlight the Hmga2-Igf2bp2 axis as a potential target for therapeutic intervention.


Assuntos
Proteínas de Ligação a DNA/genética , Proteína HMGA2/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas de Ligação a RNA/genética , Animais , Diferenciação Celular/genética , Dioxigenases , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , MicroRNAs/genética , Síndromes Mielodisplásicas/patologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Complexo Repressor Polycomb 2/genética
11.
Cancer Res ; 80(12): 2523-2536, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32341038

RESUMO

RUNX3, a RUNX family transcription factor, regulates normal hematopoiesis and functions as a tumor suppressor in various tumors in humans and mice. However, emerging studies have documented increased expression of RUNX3 in hematopoietic stem/progenitor cells (HSPC) of a subset of patients with myelodysplastic syndrome (MDS) showing a worse outcome, suggesting an oncogenic function for RUNX3 in the pathogenesis of hematologic malignancies. To elucidate the oncogenic function of RUNX3 in the pathogenesis of MDS in vivo, we generated a RUNX3-expressing, Tet2-deficient mouse model with the pancytopenia and dysplastic blood cells characteristic of MDS in patients. RUNX3-expressing cells markedly suppressed the expression levels of Runx1, a critical regulator of hemaotpoiesis in normal and malignant cells, as well as its target genes, which included crucial tumor suppressors such as Cebpa and Csf1r. RUNX3 bound these genes and remodeled their Runx1-binding regions in Tet2-deficient cells. Overexpression of RUNX3 inhibited the transcriptional function of Runx1 and compromised hematopoiesis to facilitate the development of MDS in the absence of Tet2, indicating that RUNX3 is an oncogene. Furthermore, overexpression of RUNX3 activated the transcription of Myc target genes and rendered cells sensitive to inhibition of Myc-Max heterodimerization. Collectively, these results reveal the mechanism by which RUNX3 overexpression exerts oncogenic effects on the cellular function of and transcriptional program in Tet2-deficient stem cells to drive the transformation of MDS. SIGNIFICANCE: This study defines the oncogenic effects of transcription factor RUNX3 in driving the transformation of myelodysplastic syndrome, highlighting RUNX3 as a potential target for therapeutic intervention.


Assuntos
Transformação Celular Neoplásica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Células-Tronco Hematopoéticas/patologia , Síndromes Mielodisplásicas/patologia , Animais , Medula Óssea/patologia , Proteínas de Ligação a DNA/genética , Dioxigenases , Modelos Animais de Doenças , Humanos , Células Jurkat , Camundongos , Camundongos Knockout , Síndromes Mielodisplásicas/genética , Cultura Primária de Células , Proteínas Proto-Oncogênicas/genética , Transcrição Gênica
12.
Blood ; 136(1): 106-118, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32219445

RESUMO

Mutations in JAK2, myeloproliferative leukemia virus (MPL), or calreticulin (CALR) occur in hematopoietic stem cells (HSCs) and are detected in more than 80% of patients with myeloproliferative neoplasms (MPNs). They are thought to play a driver role in MPN pathogenesis via autosomal activation of the JAK-STAT signaling cascade. Mutant CALR binds to MPL, activates downstream MPL signaling cascades, and induces essential thrombocythemia in mice. However, embryonic lethality of Calr-deficient mice precludes determination of a role for CALR in hematopoiesis. To clarify the role of CALR in normal hematopoiesis and MPN pathogenesis, we generated hematopoietic cell-specific Calr-deficient mice. CALR deficiency had little effect on the leukocyte count, hemoglobin levels, or platelet count in peripheral blood. However, Calr-deficient mice showed some hematopoietic properties of MPN, including decreased erythropoiesis and increased myeloid progenitor cells in the bone marrow and extramedullary hematopoiesis in the spleen. Transplantation experiments revealed that Calr haploinsufficiency promoted the self-renewal capacity of HSCs. We generated CALRdel52 mutant transgenic mice with Calr haploinsufficiency as a model that mimics human MPN patients and found that Calr haploinsufficiency restored the self-renewal capacity of HSCs damaged by CALR mutations. Only recipient mice transplanted with Lineage-Sca1+c-kit+ cells harboring both CALR mutation and Calr haploinsufficiency developed MPN in competitive conditions, showing that CALR haploinsufficiency was necessary for the onset of CALR-mutated MPNs.


Assuntos
Calreticulina/fisiologia , Transtornos Mieloproliferativos/etiologia , Células-Tronco/patologia , Animais , Medula Óssea/patologia , Calreticulina/deficiência , Calreticulina/genética , Autorrenovação Celular , Eritropoese , Genótipo , Hematopoese Extramedular , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Transgênicos , Transtornos Mieloproliferativos/patologia , Células-Tronco Neoplásicas/patologia , Deleção de Sequência , Transcriptoma
14.
Nat Commun ; 10(1): 1653, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30971697

RESUMO

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive subtype of acute leukemia, the cell of origin of which is considered to be precursors of plasmacytoid dendritic cells (pDCs). Since translocation (6;8)(p21;q24) is a recurrent anomaly for BPDCN, we demonstrate that a pDC-specific super-enhancer of RUNX2 is associated with the MYC promoter due to t(6;8). RUNX2 ensures the expression of pDC-signature genes in leukemic cells, but also confers survival and proliferative properties in BPDCN cells. Furthermore, the pDC-specific RUNX2 super-enhancer is hijacked to activate MYC in addition to RUNX2 expression, thereby promoting the proliferation of BPDCN. We also demonstrate that the transduction of MYC and RUNX2 is sufficient to initiate the transformation of BPDCN in mice lacking Tet2 and Tp53, providing a model that accurately recapitulates the aggressive human disease and gives an insight into the molecular mechanisms underlying the pathogenesis of BPDCN.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Células Dendríticas/patologia , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Proliferação de Células/genética , Cromossomos Humanos Par 6/genética , Cromossomos Humanos Par 8/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/genética , Dioxigenases , Elementos Facilitadores Genéticos/genética , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Humanos , Células Jurkat , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Translocação Genética/genética , Irradiação Corporal Total
15.
Support Care Cancer ; 25(3): 925-932, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27853929

RESUMO

PURPOSE: The adequacy of pain management for individuals with cancer who receive outpatient chemotherapy is unclear. The primary objective of this study was to assess pain prevalence and intensity in such patients. The secondary objectives included assessment of pain management with the pain management index (PMI) and exploration of predictors of inadequate pain management. METHODS: Cancer patients who received outpatient chemotherapy were enrolled. Patients were required to complete questionnaires covering demographic data and including the Brief Pain Inventory and the Distress Thermometer and Impact Thermometer. The PMI score was determined twice with an interval of at least 3 weeks. RESULTS: Of the 740 patients enrolled in the study, 524 individuals (70.8%) completed all questionnaires. Totals of 282 patients (53.8%) and 264 patients (50.4%) reported pain at baseline and follow-up, respectively, with ∼14% of patients having moderate or severe pain at each assessment. Totals of 365 patients (69.7%) at baseline and 320 patients (61.1%) at follow-up reported pain or were prescribed analgesics, with the rate of inadequate pain management for these patients being 39.7 and 51.6%, respectively. Multivariable analysis for 418 patients (79.8%) who had pain or required analgesics at baseline or follow-up (or both) revealed that the most significant predictor of inadequate pain management was depressive state. CONCLUSIONS: Pain in cancer patients receiving outpatient chemotherapy is prevalent and at risk for undertreatment. Pain management should be assessed on a regular basis and is likely to be improved by screening for depression.


Assuntos
Dor do Câncer/diagnóstico , Dor do Câncer/tratamento farmacológico , Neoplasias/tratamento farmacológico , Neoplasias/fisiopatologia , Manejo da Dor/métodos , Medição da Dor/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Analgésicos/uso terapêutico , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Pacientes Ambulatoriais , Prevalência , Inquéritos e Questionários , Adulto Jovem
16.
Cancer Res ; 71(2): 339-48, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21224354

RESUMO

Multiple myeloma (MM) is a currently incurable neoplasm of terminally differentiated B cells. The translocation and/or overexpression of c-MAF have been observed in human MM. Although c-MAF might function as an oncogene in human MM, there has been no report thus far describing the direct induction of MM by c-MAF overexpression in vivo. In this study, we have generated transgenic (TG) mice that express c-Maf specifically in the B-cell compartment. Aged c-Maf TG mice developed B-cell lymphomas with some clinical features that resembled those of MM, namely, plasma cell expansion and hyperglobulinemia. Quantitative RT-PCR analysis demonstrated that Ccnd2 and Itgb7, which are known target genes of c-Maf, were highly expressed in the lymphoma cells. This novel TG mouse model of the human MM t(14;16)(q32;q23) chromosomal translocation should serve to provide new insight into the role of c-MAF in tumorigenesis.


Assuntos
Modelos Animais de Doenças , Linfoma de Células B/genética , Mieloma Múltiplo/genética , Proteínas Proto-Oncogênicas c-maf/genética , Animais , Linfócitos B/metabolismo , Linfócitos B/fisiologia , Cromossomos Humanos Par 14 , Cromossomos Humanos Par 16 , Humanos , Hipergamaglobulinemia/genética , Hipergamaglobulinemia/metabolismo , Linfoma de Células B/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mieloma Múltiplo/metabolismo , Proteínas Proto-Oncogênicas c-maf/biossíntese , Translocação Genética
17.
Nihon Kokyuki Gakkai Zasshi ; 48(7): 524-8, 2010 Jul.
Artigo em Japonês | MEDLINE | ID: mdl-20684218

RESUMO

A 46-year-old man, who had been treated for Kimura disease, was found to have abnormal findings on a chest radiograph. Chest CT showed a mass shadow in the right lower lobe. Histological findings of a transbronchial biopsy specimen showed inflammatory cells infiltrating the alveolar septum. Inflammatory cells consisted of plasma cells, lymphocytes and eosinophils. He had had sustained eosinophilia and elevation of IgE for 8 years, and his serum IgG and IgG4 (3480 mg/dl) levels were elevated. Contrast-enhanced computed tomography showed multiple low density areas in both kidneys. Since immunohistochemical staining of the lung specimens revealed infiltration of IgG4-positive plasma cells (IgG4-positive/IgG-positive plasma cells=35%), we diagnosed IgG4-positive multiorgan lymphoproliferative syndrome. The mass shadow had disappeared after treatment with celestamine (0.25 mg betamethasone and 2 mg dexchlorpheniramine) for Kimura disease, but another solid consolidation appeared in the contralateral lower lobe 5 months later. After the steroid dosage was increased, the consolidation improved. To the best of our knowledge, this case is the first report of IgG4-positive multi-organ lymphoproliferative syndrome associated with Kimura disease.


Assuntos
Hiperplasia Angiolinfoide com Eosinofilia/complicações , Imunoglobulina G/sangue , Leucemia Linfocítica Granular Grande/complicações , Humanos , Masculino , Pessoa de Meia-Idade
18.
Nihon Kokyuki Gakkai Zasshi ; 46(6): 493-6, 2008 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-18592997

RESUMO

A 69-year-old man was admitted to our hospital with dyspnea on effort. A chest high resolution CT showed enlargement of peripheral vessels and small nodules in both lower and peripheral lung fields. We diagnosed this case as pulmonary tumor thrombotic microangiopathy, because histological findings of specimen obtained by transbronchial lung biopsy revealed microscopic tumor emboli and intimal proliferation in small pulmonary vessels. After conducting systemic examinations, it was found that the patient had gastric cancer. Generally it is difficult to diagnose pulmonary tumor thrombotic microangiopathy in patients during life. To the best of our knowledge, this is the first case of pulmonary tumor thronbotic microangiopthy diagnosed on the basis of transbronchial lung biopsy.


Assuntos
Adenocarcinoma/diagnóstico , Adenocarcinoma/secundário , Biópsia/métodos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/secundário , Pulmão/patologia , Células Neoplásicas Circulantes/patologia , Adenocarcinoma/patologia , Idoso , Humanos , Pulmão/irrigação sanguínea , Neoplasias Pulmonares/patologia , Masculino , Microcirculação/patologia , Neoplasias Gástricas/patologia , Tomografia Computadorizada por Raios X
19.
J Immunol ; 180(1): 207-13, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18097021

RESUMO

The myeloid-associated Ig-like receptor family (CD300) consists of nine activating or inhibitory cell surface receptors preferentially expressed on myeloid cells and are encoded by the genes in a small cluster on mouse chromosome 11. One of the receptors, CD300LF (MAIR-V), has a long cytoplasmic tail containing two consensus ITIMs and an immunoreceptor tyrosine-based switching motif, suggesting that CD300LF regulates the activation of myeloid cells. However, the functional characteristics of this receptor are still incompletely understood. In this study, we demonstrate that cross-linking CD300LF with anti-CD300LF mAb induced cell death in peritoneal macrophages as well as in several transfectants expressing CD300LF. CD300LF-mediated cell death was dependent on the cytoplasmic region but did not require an ITIM or immunoreceptor tyrosine-based switching motif. Scanning electron microscopy revealed a loss of blebs from the surface of the dead cells mediated by CD300LF, a morphological feature similar to that observed in apoptotic cells. However, CD300LF-mediated cell death was not inhibited by a caspase inhibitor, N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone, or autophagy inhibitors, 3-methyladenine or N-acetyl-L-cystein. Moreover, the splicing isoform of a transcription factor, X-box binding protein-1, which is produced in dead cells as a response to endoplasmic reticulum stress, was not detected. Together, these results indicate that CD300LF mediates caspase and endoplasmic reticulum stress-independent cell death by a novel mechanism.


Assuntos
Autofagia , Retículo Endoplasmático/imunologia , Macrófagos Peritoneais/imunologia , Células Mieloides/imunologia , Receptores Imunológicos/fisiologia , Acetilcisteína/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Inibidores de Caspase , Caspases/metabolismo , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/química , Células Mieloides/efeitos dos fármacos , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição
20.
Mol Immunol ; 45(1): 289-94, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17543387

RESUMO

Neutrophils play a central role in host defenses against infectious microbial pathogens. However, molecular basis for their triggering signals has been incompletely understood. Here, we show molecular and functional characteristics of myeloid-associated immunoglobulin-like receptor (MAIR)-IV. MAIR-IV was preferentially expressed on neutrophils in the peripheral blood, bone marrow, peritoneal cavity and spleen. MAIR-IV physically associates with immunoreceptor tyrosine based activating motif (ITAM)-bearing adaptor FcRgamma chain. Cross-linking MAIR-IV with anti-MAIR-IV monoclonal antibody significantly induced secretion of proinflammatory cytokines TNF-alpha and IL-6 from neutrophils. Thus, MAIR-IV may regulate activation of neutrophils and play an important role for innate immunity.


Assuntos
Ativação de Neutrófilo/imunologia , Receptores de Imunoglobulina Polimérica/imunologia , Animais , Cromossomos de Mamíferos , Citocinas/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Neutrófilos/metabolismo , Ligação Proteica , Receptores de IgG/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA