Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Int J Mol Sci ; 24(11)2023 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-37298318

RESUMEN

Turmeric (Curcuma longa) contains various compounds that potentially improve health. Bisacurone is a turmeric-derived compound but has been less studied compared to other compounds, such as curcumin. In this study, we aimed to evaluate the anti-inflammatory and lipid-lowering effects of bisacurone in high-fat diet (HFD)-fed mice. Mice were fed HFD to induce lipidemia and orally administered bisacurone daily for two weeks. Bisacurone reduced liver weight, serum cholesterol and triglyceride levels, and blood viscosity in mice. Splenocytes from bisacurone-treated mice produced lower levels of the pro-inflammatory cytokines IL-6 and TNF-α upon stimulation with a toll-like receptor (TLR) 4 ligand, lipopolysaccharide (LPS), and TLR1/2 ligand, Pam3CSK4, than those from untreated mice. Bisacurone also inhibited LPS-induced IL-6 and TNF-α production in the murine macrophage cell line, RAW264.7. Western blot analysis revealed that bisacurone inhibited the phosphorylation of IKKα/ß and NF-κB p65 subunit, but not of the mitogen-activated protein kinases, p38 kinase and p42/44 kinases, and c-Jun N-terminal kinase in the cells. Collectively, these results suggest that bisacurone has the potential to reduce serum lipid levels and blood viscosity in mice with high-fat diet-induced lipidemia and modulate inflammation via inhibition of NF-κB-mediated pathways.


Asunto(s)
Curcuma , FN-kappa B , Animales , Ratones , FN-kappa B/metabolismo , Curcuma/metabolismo , Factor de Necrosis Tumoral alfa , Lipopolisacáridos/farmacología , Dieta Alta en Grasa/efectos adversos , Ligandos , Interleucina-6 , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Citocinas/metabolismo
2.
Kidney Int ; 101(1): 92-105, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34767829

RESUMEN

Space travel burdens health by imposing considerable environmental stress associated with radioactivity and microgravity. In particular, gravity change predominantly impacts blood pressure and bone homeostasis, both of which are controlled mainly by the kidneys. Nuclear factor erythroid-2-related transcription factor 2 (Nrf2) plays essential roles in protecting the kidneys from various environmental stresses and injuries. To elucidate the effects of space travel on mammals in preparation for the upcoming space era, our study investigated the contribution of Nrf2 to kidney function in mice two days after their return from a 31-day stay in the International Space Station using Nrf2 knockout mice. Meaningfully, expression levels of genes regulating bone mineralization, blood pressure and lipid metabolism were found to be significantly altered in the kidneys after space travel in an Nrf2-independent manner. In particular, uridine diphosphate-glucuronosyltransferase 1A (Ugt1a) isoform genes were found to be expressed in an Nrf2-dependent manner and induced exclusively in the kidneys after return to Earth. Since spaceflight elevated the concentrations of fatty acids in the mouse plasma, we suggest that Ugt1a isoform expression in the kidneys was induced to promote glucuronidation of excessively accumulated lipids and excrete them into urine after the return from space. Thus, the kidneys were proven to play central roles in adaptation to gravity changes caused by going to and returning from space by controlling blood pressure and bone mineralization. Additionally, kidney Ugt1a isoform induction after space travel implies a significant role of the kidneys for space travelers in the excretion of excessive lipids.


Asunto(s)
Metabolismo de los Lípidos , Vuelo Espacial , Animales , Presión Sanguínea/genética , Calcificación Fisiológica , Expresión Génica , Riñón/metabolismo , Metabolismo de los Lípidos/genética , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo
3.
Cancer Sci ; 112(6): 2454-2466, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33759313

RESUMEN

The use of patient-derived xenografts (PDXs) has recently attracted attention as a drug discovery platform with a high predictive clinical efficacy and a preserved tumor heterogeneity. Given the racial differences in genetic variations, it would be desirable to establish a PDX library from Japanese cancer patients on a large scale. We thus tried to construct the Japanese PDX (J-PDX) library with a detailed clinical information for further clinical utilization. Between August 2018 and May 2020, a total of 1126 cancer specimens from 1079 patients were obtained at the National Cancer Center Hospital and National Cancer Center Hospital East, Japan, and were immediately transplanted to immunodeficient mice at the National Cancer Center Research Institute. A total of 298 cross-cancer PDXs were successfully established. The time to engraftment varied greatly by cancer subtypes, especially in the first passage. The engraftment rate was strongly affected by the clinical stage and survival time of the original patients. Approximately 1 year was needed from tumor collection to the time when coclinical trials were conducted to test the clinical utility. The 1-year survival rates of the patients who were involved in establishing the PDX differed significantly, from 95.6% for colorectal cancer to 56.3% for lung cancer. The J-PDX library consisting of a wide range of cancer subtypes has been successfully established as a platform for drug discovery and development in Japan. When conducting coclinical trials, it is necessary to consider the target cancer type, stage, and engraftment rate in light of this report.


Asunto(s)
Neoplasias/mortalidad , Neoplasias/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Femenino , Humanos , Japón/etnología , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Trasplante de Neoplasias , Especificidad de Órganos , Modelación Específica para el Paciente , Adulto Joven
4.
Cancer Sci ; 111(2): 667-678, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31828882

RESUMEN

Aberrant activation of NRF2 is as a critical prognostic factor that drives the malignant progression of various cancers. Cancer cells with persistent NRF2 activation heavily rely on NRF2 activity for therapeutic resistance and aggressive tumorigenic capacity. To clarify the metabolic features of NRF2-activated lung cancers, we conducted targeted metabolomic (T-Met) and global metabolomic (G-Met) analyses of non-small-cell lung cancer (NSCLC) cell lines in combination with exome and transcriptome analyses. Exome analysis of 88 cell lines (49 adenocarcinoma, 14 large cell carcinoma, 15 squamous cell carcinoma and 10 others) identified non-synonymous mutations in the KEAP1, NRF2 and CUL3 genes. Judging from the elevated expression of NRF2 target genes, these mutations are expected to result in the constitutive stabilization of NRF2. Out of the 88 cell lines, 52 NSCLC cell lines (29 adenocarcinoma, 10 large cell carcinoma, 9 squamous cell carcinoma and 4 others) were subjected to T-Met analysis. Classification of the 52 cell lines into three groups according to the NRF2 target gene expression enabled us to draw typical metabolomic signatures induced by NRF2 activation. From the 52 cell lines, 18 NSCLC cell lines (14 adenocarcinoma, 2 large cell carcinoma, 1 squamous cell carcinoma and 1 others) were further chosen for G-Met and detailed transcriptome analyses. G-Met analysis of their culture supernatants revealed novel metabolites associated with NRF2 activity, which may be potential diagnostic biomarkers of NRF2 activation. This study also provides useful information for the exploration of new metabolic nodes for selective toxicity towards NRF2-activated NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Metabolómica/métodos , Mutación , Factor 2 Relacionado con NF-E2/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proteínas Cullin/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Neoplasias Pulmonares/genética , Factor 2 Relacionado con NF-E2/genética , Secuenciación del Exoma
5.
IUBMB Life ; 72(1): 159-169, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31820561

RESUMEN

Chromosomal inversion and translocation between 3q21 and 3q26 [inv (3)(q21.3q26.2) and t(3;3)(q21.3;q26.2), respectively] give rise to acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), which have poor prognoses. The chromosomal rearrangements reposition a GATA2 distal hematopoietic enhancer from the original 3q21 locus to the EVI1 (also known as MECOM) locus on 3q26. Therefore, the GATA2 enhancer from one of two GATA2 alleles drives EVI1 gene expression in hematopoietic stem and progenitor cells, which promotes the accumulation of abnormal progenitors and induces leukemogenesis. On the other hand, one allele of the GATA2 gene loses its enhancer, which results in reduced GATA2 expression. The GATA2 gene encodes a transcription factor critical for the generation and maintenance of hematopoietic stem and progenitor cells. GATA2 haploinsufficiency has been known to cause immunodeficiency and myeloid leukemia. Notably, reduced GATA2 expression suppresses the differentiation but promotes the proliferation of EVI1-expressing leukemic cells, which accelerates EVI1-driven leukemogenesis. A series of studies have shown that the GATA2 enhancer repositioning caused by the chromosomal rearrangements between 3q21 and 3q26 provokes misexpression of both the EVI1 and GATA2 genes and that these two effects coordinately elicit high-risk leukemia.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 3/genética , Elementos de Facilitación Genéticos , Factor de Transcripción GATA2/genética , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Factor de Transcripción GATA2/metabolismo , Reordenamiento Génico , Humanos , Translocación Genética
6.
Blood ; 130(7): 908-919, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28630119

RESUMEN

Chromosomal rearrangements between 3q21 and 3q26 induce inappropriate EVI1 expression by recruiting a GATA2-distal hematopoietic enhancer (G2DHE) to the proximity of the EVI1 gene, leading to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). The acquisition of G2DHE by the EVI1 gene reciprocally deprives this enhancer of 1 of the 2 GATA2 alleles, resulting in a loss-of-function genetic reduction in GATA2 abundance. Because GATA2 haploinsufficiency is strongly associated with MDS and AML, we asked whether EVI1 misexpression and GATA2 haploinsufficiency both contributed to the observed leukemogenesis by using a 3q21q26 mouse model that recapitulates the G2DHE-driven EVI1 misexpression, but in this case, it was coupled to a Gata2 heterozygous germ line deletion. Of note, the Gata2 heterozygous deletion promoted the EVI1-provoked leukemic transformation, resulting in early onset of leukemia. The 3q21q26 mice suffered from leukemia in which B220+ cells and/or Gr1+ leukemic cells occupied their bone marrows. We found that the B220+Gr1-c-Kit+ population contained leukemia-initiating cells and supplied Gr1+ leukemia cells in the 3q21q26 leukemia. When Gata2 expression levels in the B220+Gr1-c-Kit+ cells were decreased as a result of Gata2 heterozygous deletion or spontaneous phenomenon, myeloid differentiation of the B220+Gr1-c-Kit+ cells was suppressed, and the cells acquired induced proliferation as well as B-lymphoid-primed characteristics. Competitive transplantation analysis revealed that Gata2 heterozygous deletion confers selective advantage to EVI1-expressing leukemia cell expansion in recipient mice. These results demonstrate that both the inappropriate stimulation of EVI1 and the loss of 1 allele equivalent of Gata2 expression contribute to the acceleration of leukemogenesis.


Asunto(s)
Carcinogénesis/patología , Proteínas de Unión al ADN/metabolismo , Factor de Transcripción GATA2/genética , Haploinsuficiencia/genética , Leucemia/patología , Factores de Transcripción/metabolismo , Alelos , Animales , Carcinogénesis/genética , Diferenciación Celular , Proliferación Celular , Cromosomas de los Mamíferos/genética , Metabolismo Energético/genética , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Leucemia/genética , Proteína del Locus del Complejo MDS1 y EV11 , Ratones Endogámicos C57BL , Modelos Biológicos , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proto-Oncogenes , Estrés Fisiológico/genética
7.
Proc Natl Acad Sci U S A ; 112(39): 12169-74, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26371321

RESUMEN

Sickle cell disease (SCD) is an inherited disorder caused by a point mutation in the ß-globin gene, leading to the production of abnormally shaped red blood cells. Sickle cells are prone to hemolysis and thereby release free heme into plasma, causing oxidative stress and inflammation that in turn result in damage to multiple organs. The transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) is a master regulator of the antioxidant cell-defense system. Here we show that constitutive Nrf2 activation by ablation of its negative regulator Keap1 (kelch-like ECH-associated protein 1) significantly improves symptoms in SCD model mice. SCD mice exhibit severe liver damage and lung inflammation associated with high expression levels of proinflammatory cytokines and adhesion molecules compared with normal mice. Importantly, these symptoms subsided after Nrf2 activation. Although hemolysis and stress erythropoiesis did not change substantially in the Nrf2-activated SCD mice, Nrf2 promoted the elimination of plasma heme released by sickle cells' hemolysis and thereby reduced oxidative stress and inflammation, demonstrating that Nrf2 activation reduces organ damage and segregates inflammation from prevention of hemolysis in SCD mice. Furthermore, administration of the Nrf2 inducer CDDO-Im (2-cyano-3, 12 dioxooleana-1, 9 diene-28-imidazolide) also relieved inflammation and organ failure in SCD mice. These results support the contention that Nrf2 induction may be an important means to protect organs from the pathophysiology of sickle cell-induced damage.


Asunto(s)
Anemia de Células Falciformes/complicaciones , Inflamación/genética , Inflamación/terapia , Hígado/patología , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/genética , Activación Transcripcional/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Anemia de Células Falciformes/terapia , Animales , Proteínas del Citoesqueleto/genética , Cartilla de ADN/genética , Citometría de Flujo , Técnicas de Inactivación de Genes , Immunoblotting , Proteína 1 Asociada A ECH Tipo Kelch , Luciferasas , Ratones , Factor 2 Relacionado con NF-E2/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Rinsho Ketsueki ; 58(7): 806-812, 2017.
Artículo en Japonés | MEDLINE | ID: mdl-28781278

RESUMEN

Chromosomal rearrangements between 3q21 and 3q26 induce the abnormal expression of the EVI1 gene on 3q26, which results in leukemia and a poor prognosis. In the rearranged allele, we found that the GATA2 gene enhancer on 3q21 localizes in close proximity to the EVI1 gene. To examine the contribution of the GATA2 gene enhancer upon the abnormal expression of EVI1 and leukemogenesis, we established a leukemia mouse model (3q21q26 mouse) harboring a transgene recapitulating a 196-kb inverted allele between 3q21 and 3q26 by linking two bacterial artificial chromosome clones. The 3q21q26 mice demonstrated high EVI1 transgene expression specifically in hematopoietic progenitors and developed leukemia after 6 months of age. Of note, by deleting the GATA2 enhancer, EVI1 transgene expression and leukemogenesis were significantly suppressed, indicating that the GATA2 enhancer drives the abnormal expression of EVI1 in the rearranged allele and induces leukemogenesis. While the EVI1 gene gains the GATA2 enhancer, GATA2 gene loses its enhancer. Therefore, GATA2 expression levels are reduced in leukemic cells with such chromosomal rearrangements. To examine the contribution of GATA2 heterozygous deletion upon leukemogenesis, we crossed the 3q21q26 mice with Gata2 heterozygous knockout mice to generate compound mutant mice recapitulating both abnormal EVI1 expression and Gata2 heterozygous deletion. The compound mutant mice developed leukemia earlier than the 3q21q26 mice did. These results indicate that GATA2 heterozygous deletion accelerates leukemogenesis driven by the abnormal expression of EVI1.


Asunto(s)
Cromosomas Humanos Par 3 , Factor de Transcripción GATA2/genética , Reordenamiento Génico , Proteína del Locus del Complejo MDS1 y EV11/genética , Animales , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia/genética , Proteína del Locus del Complejo MDS1 y EV11/metabolismo
9.
Blood ; 122(20): 3450-60, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24021675

RESUMEN

GATA1 is a master regulator of hematopoietic differentiation, but Gata1 expression is inactivated in hematopoietic stem cells (HSCs). Using a bacterial artificial chromosome containing the Gata1 gene modified with green fluorescent protein (GFP) reporter, we explored the function of the 3.7-kb Gata1 upstream region (GdC region) that harbors 3 core cis-elements: Gata1 hematopoietic enhancer, double GATA-motif, and CACCC-motif. Transgenic GFP expression directed by the Gata1-BAC faithfully recapitulated the endogenous Gata1 expression pattern. However, deletion of the GdC-region eliminated reporter expression in all hematopoietic cells. To test whether the combination of the core cis-elements represents the regulatory function of the GdC-region, we replaced the region with a 659-bp minigene that linked the three cis-elements (MG-GFP). The GFP reporter expression directed by the MG-GFP BAC fully recapitulated the erythroid-megakaryocytic Gata1 expression. However, the GFP expression was aberrantly increased in the HSCs and was associated with decreases in DNA methylation and abundant GATA2 binding to the transgenic MG-GFP allele. The 3.2-kb sequences interspaced between the Gata1 hematopoietic enhancer and the double GATA-motif were able to recruit DNA methyltransferase 1, thereby exerting a cis-repressive function in the HSC-like cell line. These results indicate that the 3.2-kb interspacing sequences inactivate Gata1 by maintaining DNA-methylation in the HSCs.


Asunto(s)
Células Eritroides/metabolismo , Factor de Transcripción GATA1/genética , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Hematopoyéticas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Linaje de la Célula , Células Cultivadas/metabolismo , Cromosomas Artificiales Bacterianos , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Elementos de Facilitación Genéticos/genética , Eritropoyesis/genética , Factor de Transcripción GATA1/fisiología , Factor de Transcripción GATA2/metabolismo , Silenciador del Gen , Genes Reporteros , Genes Sintéticos , Hígado/citología , Hígado/embriología , Megacariocitos/metabolismo , Ratones , Ratones Transgénicos , Motivos de Nucleótidos/genética , Eliminación de Secuencia , Activación Transcripcional/genética
10.
Blood ; 121(16): 3181-4, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23440243

RESUMEN

Children with Down syndrome have an increased incidence of transient abnormal myelopoiesis (TAM) and acute megakaryoblastic leukemia. The majority of these cases harbor somatic mutations in the GATA1 gene, which results in the loss of full-length GATA1. Only a truncated isoform of GATA1 that lacks the N-terminal 83 amino acids (GATA1-S) remains. We found through genetic studies of 106 patients with TAM that internally deleted GATA1 proteins (GATA1-IDs) lacking amino acid residues 77-119 or 74-88 (created by splicing mutations) contributed to the genesis of TAM in 6 patients. Analyses of GATA1-deficient embryonic megakaryocytic progenitors revealed that the GATA1 function in growth restriction was disrupted in GATA1-IDs. In contrast, GATA1-S promoted megakaryocyte proliferation more profoundly than that induced by GATA1 deficiency. These results indicate that the internally deleted regions play important roles in megakaryocyte proliferation and that perturbation of this mechanism is involved in the pathogenesis of TAM.


Asunto(s)
Síndrome de Down/sangre , Síndrome de Down/complicaciones , Síndrome de Down/genética , Factor de Transcripción GATA1/genética , Reacción Leucemoide/complicaciones , Reacción Leucemoide/genética , Eliminación de Secuencia , Proliferación Celular , Niño , Síndrome de Down/patología , Humanos , Reacción Leucemoide/patología , Megacariocitos/metabolismo , Megacariocitos/patología
11.
Genes Cells ; 18(10): 886-98, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23890289

RESUMEN

GATA1 is a transcription factor essential for erythropoiesis and megakaryopoiesis. It has been found that Gata1 gene knockdown heterozygous female (Gata1(G1.05/+)) mice spontaneously develop erythroblastic leukemias. In this study, we have generated a novel Gata1 knockdown erythroblastic cell line, designated GAK14, from the leukemia cells in the Gata1(G1.05/+) mice. Although GAK14 cells maintain immature phenotype on OP9 stromal cells in the presence of erythropoietin and stem cell factor, the cells produce Gr-1-, Mac1-, B220-, CD3e- or CD49b-positive hematopoietic cells when co-cultured with DAS104-8 feeder cells. However, GAK14 cells did not produce erythroid and megakaryocytic lineages, perhaps due to the absence of GATA1. Indeed, GAK14 cells became capable of differentiating into mature erythroid cells when complemented with full-length GATA1 and co-cultured with fetal liver-derived FLS5 stromal cells. This differentiation potential was impaired when GATA1 lacking the N-terminal domain was complemented. The N-terminal domain is known to contribute to the pathogenesis of transient abnormal myelopoiesis and acute megakaryoblastic leukemia related to Down syndrome. These results thus showed that GAK14 cells will serve as a powerful tool for dissecting domain function of GATA1 and that the GATA1 N-terminal domain is essential for the erythroid differentiation of GAK14 cells.


Asunto(s)
Aminoácidos/genética , Línea Celular Tumoral , Células Precursoras Eritroides/fisiología , Eritropoyesis , Factor de Transcripción GATA1/química , Factor de Transcripción GATA1/metabolismo , Leucemia Eritroblástica Aguda , Animales , Linaje de la Célula , Técnicas de Cocultivo , Células Precursoras Eritroides/metabolismo , Femenino , Factor de Transcripción GATA1/genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Células Progenitoras de Megacariocitos/fisiología , Ratones , Mielopoyesis , Estructura Terciaria de Proteína
12.
Genes Cells ; 18(11): 921-33, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23911012

RESUMEN

Transcription factor GATA2 is highly expressed in hematopoietic stem cells and progenitors, whereas its expression declines after erythroid commitment of progenitors. In contrast, the start of GATA1 expression coincides with the erythroid commitment and increases along with the erythroid differentiation. We refer this dynamic transition of GATA factor expression to as the 'GATA factor switching'. Here, we examined contribution of the GATA factor switching to the erythroid differentiation. In Gata1-knockdown embryos that concomitantly express Gata2-GFP reporter, high-level expression of GFP reporter was detected in accumulated immature hematopoietic cells with impaired differentiation, demonstrating that GATA1 represses Gata2 gene expression in hematopoietic progenitors in vivo. We have conducted chromatin immunoprecipitation (ChIP) on microarray analyses of GATA2 and GATA1, and results indicate that the GATA1-binding sites widely overlap with the sites pre-occupied by GATA2 before the GATA1 expression. Importantly, erythroid genes harboring GATA boxes bound by both GATA1 and GATA2 tend to be expressed in immature erythroid cells, whereas those harboring GATA boxes to which GATA1 binds highly but GATA2 binds only weakly are important for the mature erythroid cell function. Our results thus support the contention that preceding binding of GATA2 helps the following binding of GATA1 and thereby secures smooth expression of the transient-phase genes.


Asunto(s)
Células Eritroides/citología , Eritropoyesis/fisiología , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA2/genética , Células Madre Hematopoyéticas/citología , Animales , Sitios de Unión , Diferenciación Celular , Células Eritroides/metabolismo , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Transgénicos
13.
Artículo en Inglés | MEDLINE | ID: mdl-38512455

RESUMEN

OBJECTIVE: We examined cases in which delirium developed after thoracic surgery under general anesthesia at our hospital to determine the predictive factors for postoperative delirium, as well as the perioperative findings in cases showing postoperative delirium. METHODS: This retrospective study included 1674 patients who underwent surgery under general anesthesia at our hospital between 2012 and 2022, A psychiatrist diagnosed postoperative delirium using the Confusion Assessment Method. RESULTS: There were 99 (5.9%) patients with postoperative delirium in our study, including 85 (86%) men, of whom 31 (31%) had a history of cerebrovascular disease. The incidence of postoperative delirium in patients aged > 80 years was 20% (36/182). The postoperative delirium group showed significantly longer hospital stays and more frequent postoperative complications than the group without postoperative delirium. In univariate analysis, age ≥ 80 years, male sex, history of cerebrovascular disease, hypertension, history of atrial fibrillation, and history of smoking were identified as significant factors, while multivariate analysis identified age ≥ 80 years, male sex, history of cerebrovascular disease, hypertension, and history of smoking as significant factors (odds ratios = 5.15, 2.04, 3.10, 1.67, and 2.36, respectively). In the 169 cases with none of these five factors, the postoperative delirium risk was 0% (0/169). CONCLUSIONS: In patients undergoing thoracic surgery, predictive factors for postoperative delirium include age ≥ 80 years, male sex, history of cerebrovascular disease, hypertension, and smoking history. The findings also indicate that patients with these risk factors may require psychiatric consultation before surgery.

14.
Cell Rep Med ; 5(5): 101532, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38670097

RESUMEN

Ovarian clear cell carcinoma (OCCC) is a gynecological cancer with a dismal prognosis; however, the mechanism underlying OCCC chemoresistance is not well understood. To explore the intracellular networks associated with the chemoresistance, we analyze surgical specimens by performing integrative analyses that combine single-cell analyses and spatial transcriptomics. We find that a chemoresistant OCCC subpopulation with elevated HIF activity localizes mainly in areas populated by cancer-associated fibroblasts (CAFs) with a myofibroblastic phenotype, which is corroborated by quantitative immunostaining. CAF-enhanced chemoresistance and HIF-1α induction are recapitulated in co-culture assays, which show that cancer-derived platelet-derived growth factor (PDGF) contributes to the chemoresistance and HIF-1α induction via PDGF receptor signaling in CAFs. Ripretinib is identified as an effective receptor tyrosine kinase inhibitor against CAF survival. In the co-culture system and xenograft tumors, ripretinib prevents CAF survival and suppresses OCCC proliferation in the presence of carboplatin, indicating that combination of conventional chemotherapy and CAF-targeted agents is effective against OCCC.


Asunto(s)
Fibroblastos Asociados al Cáncer , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias Ováricas , Factor de Crecimiento Derivado de Plaquetas , Transducción de Señal , Femenino , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Ratones , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Progresión de la Enfermedad , Técnicas de Cocultivo , Proliferación Celular/efectos de los fármacos , Ratones Desnudos , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Adenocarcinoma de Células Claras/tratamiento farmacológico , Adenocarcinoma de Células Claras/genética , Retroalimentación Fisiológica/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Commun Biol ; 6(1): 875, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37626149

RESUMEN

Spaceflight-related stresses impact health via various body systems, including the haematopoietic and immune systems, with effects ranging from moderate alterations of homoeostasis to serious illness. Oxidative stress appears to be involved in these changes, and the transcription factor Nrf2, which regulates expression of a set of cytoprotective and antioxidative stress response genes, has been implicated in the response to spaceflight-induced stresses. Here, we show through analyses of mice from the MHU-3 project, in which Nrf2-knockout mice travelled in space for 31 days, that mice lacking Nrf2 suffer more seriously from spaceflight-induced immunosuppression than wild-type mice. We discovered that a one-month spaceflight-triggered the expression of tissue inflammatory marker genes in wild-type mice, an effect that was even more pronounced in the absence of Nrf2. Concomitant with induction of inflammatory conditions, the consumption of coagulation-fibrinolytic factors and platelets was elevated by spaceflight and further accelerated by Nrf2 deficiency. These results highlight that Nrf2 mitigates spaceflight-induced inflammation, subsequent immunosuppression, and thrombotic microangiopathy. These observations reveal a new strategy to relieve health problems encountered during spaceflight.


Asunto(s)
Vuelo Espacial , Microangiopatías Trombóticas , Animales , Ratones , Terapia de Inmunosupresión , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética
16.
Cell Mol Gastroenterol Hepatol ; 15(1): 153-178, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36115578

RESUMEN

BACKGROUND & AIMS: NF-E2-related factor 2 (NRF2) is a transcription factor that regulates cytoprotective gene expression in response to oxidative and electrophilic stresses. NRF2 activity is mainly controlled by Kelch-like ECH-associated protein 1 (KEAP1). Constitutive NRF2 activation by NRF2 mutations or KEAP1 dysfunction results in a poor prognosis for esophageal squamous cell carcinoma (ESCC) through the activation of cytoprotective functions. However, the detailed contributions of NRF2 to ESCC initiation or promotion have not been clarified. Here, we investigated the fate of NRF2-activated cells in the esophageal epithelium. METHODS: We generated tamoxifen-inducible, squamous epithelium-specific Keap1 conditional knockout (Keap1-cKO) mice in which NRF2 was inducibly activated in a subset of cells at the adult stage. Histologic, quantitative reverse-transcription polymerase chain reaction, single-cell RNA-sequencing, and carcinogen experiments were conducted to analyze the Keap1-cKO esophagus. RESULTS: KEAP1-deleted/NRF2-activated cells and cells with normal NRF2 expression (KEAP1-normal cells) coexisted in the Keap1-cKO esophageal epithelium in approximately equal numbers, and NRF2-activated cells formed dysplastic lesions. NRF2-activated cells exhibited weaker attachment to the basement membrane and gradually disappeared from the epithelium. In contrast, neighboring KEAP1-normal cells exhibited accelerated proliferation and started dominating the epithelium but accumulated DNA damage that triggered carcinogenesis upon carcinogen exposure. CONCLUSIONS: Constitutive NRF2 activation promotes the selective elimination of epithelial cells via cell competition, but this competition induces DNA damage in neighboring KEAP1-normal cells, which predisposes them to chemical-induced ESCC.


Asunto(s)
Epitelio , Factor 2 Relacionado con NF-E2 , Animales , Ratones , Carcinógenos , Epitelio/patología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Esófago/patología
17.
Clin Cancer Res ; 29(12): 2239-2249, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-36976261

RESUMEN

PURPOSE: Uterine carcinosarcoma (UCS), a subtype of endometrial carcinoma, is a rare and aggressive cancer with a poor prognosis. High clinical efficacy of trastuzumab deruxtecan (T-DXd) in HER2-expressing UCS was recently reported in a phase II trial (STATICE trial). We performed a co-clinical study of T-DXd using patient-derived xenograft (PDX) models of participants in the STATICE trial. EXPERIMENTAL DESIGN: Tumor specimens were resected during primary surgery or biopsied at recurrence from patients with UCS and transplanted into immunodeficient mice. Seven UCS-PDXs from six patients were established and HER2, estrogen receptor (ER), and p53 expression in PDX and the original tumor was assessed. Drug efficacy tests were performed using six of the seven PDXs. Of the six UCS-PDXs tested, two were derived from patients enrolled in the STATICE trial. RESULTS: The histopathological characteristics of the six PDXs were well-conserved from the original tumors. HER2 expression was 1+ in all PDXs, and ER and p53 expression was almost similar to that in the original tumors. Remarkable tumor shrinkage after T-DXd administration was observed in four of the six PDXs (67%), comparable with the response rate (70%) of HER2 1+ patients in the STATICE trial. Two patients enrolled in the STATICE trial showed partial response as the best response, and the clinical effect was well-replicated with marked tumor shrinkage. CONCLUSIONS: We successfully performed a co-clinical study of T-DXd in HER2-expressing UCS, along with the STATICE trial. Our PDX models can predict clinical efficacy and serve as an effective preclinical evaluation platform.


Asunto(s)
Carcinosarcoma , Inmunoconjugados , Humanos , Animales , Ratones , Receptor ErbB-2/metabolismo , Xenoinjertos , Proteína p53 Supresora de Tumor , Trastuzumab/metabolismo , Camptotecina , Inmunoconjugados/metabolismo , Resultado del Tratamiento , Receptores de Estrógenos/metabolismo
18.
Antioxidants (Basel) ; 11(3)2022 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-35326187

RESUMEN

In a multicellular environment, many different types of cells interact with each other. The KEAP1-NRF2 system defends against electrophilic and oxidative stresses in various types of cells. However, the KEAP1-NRF2 system also regulates the expression of genes involved in cell proliferation and inflammation, indicating that the system plays cell type-specific roles. In this review, we introduce the multifarious roles of the KEAP1-NRF2 system in various types of cells, especially focusing on cancer and inflammatory diseases. Cancer cells frequently hijack the KEAP1-NRF2 system, and NRF2 activation confers cancer cells with a proliferative advantage and therapeutic resistance. In contrast, the activation of NRF2 in immune cells, especially in myeloid cells, suppresses tumor development. In chronic inflammatory diseases, such as sickle cell disease, NRF2 activation in myeloid and endothelial cells represses the expression of proinflammatory cytokine and adherent molecule genes, mitigating inflammation and organ damage. Based on these cell-specific roles played by the KEAP1-NRF2 system, NRF2 inducers have been utilized for the treatment of inflammatory diseases. In addition, the use of NRF2 inducers and/or inhibitors with canonical antineoplastic drugs is an emerging approach to cancer treatment.

19.
Mol Cell Biol ; 42(7): e0056321, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35674451

RESUMEN

Nrf2 activates cytoprotective gene expression, and Nrf2 activity is regulated through at least two protein degradation pathways: the Keap1-mediated and ß-TrCP-mediated pathways. To address the relative contributions of these pathways, we generated knock-in mouse lines expressing an Nrf2SA mutant that harbored two substitution mutations of serine residues interacting with ß-TrCP. The homozygous (Nrf2SA/SA) mice grew normally, with Nrf2 levels comparable to those of wild-type (WT) mice under unstressed conditions. However, when Keap1 activity was suppressed, high levels of Nrf2 accumulated in Nrf2SA/SA macrophages compared with that in WT macrophages. We crossed Nrf2SA/SA mice with mice in which Keap1 was knocked down to two different levels. We found that the Nrf2SA/SA mutation induced higher Nrf2 activity when the Keap1 level was strongly reduced, and these mice showed severe growth retardation. However, activation and growth retardation were not evident when Keap1 was moderately suppressed. These increases in Nrf2 activity induced by the Nrf2SA mutation caused severe hyperplasia and hyperkeratosis in the esophageal epithelium but did not cause abnormalities in the other tissues/organs examined. These results indicate that the ß-TrCP-mediated pathway cooperates with the Keap1-mediated pathway to regulate Nrf2 activity, which is apparent when the Keap1-mediated pathway is profoundly suppressed.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Proteínas con Repetición de beta-Transducina , Animales , Trastornos del Crecimiento , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Proteínas con Repetición de beta-Transducina/química
20.
Free Radic Biol Med ; 187: 92-104, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35618180

RESUMEN

The Keap1-Nrf2 system is the master regulator of the cellular response against oxidative and xenobiotic stresses. Constitutive activation of Nrf2 is frequently observed in various types of cancers. Nrf2 hyperactivation induces metabolic reprogramming in cancer cells, which supports the increased energy demand required for rapid proliferation and confers high-level resistance against anticancer radio/chemotherapy. Hence, Nrf2 inhibition has emerged as an attractive therapeutic strategy to counter such acquired resistance in Nrf2-activated tumors. We previously identified Halofuginone (HF) as a promising Nrf2 inhibitor. In this study, we pursued preclinical characterization of HF and found that while HF markedly reduced the viability of cancer cells, it also caused severe hematopoietic and immune cell suppression in a dose-dependent manner. Hence, to overcome this toxicity, we decided to employ a nanomedicine approach to HF. We found that encapsulation of HF into a polymeric micelle (HF micelle; HFm) largely relieved the systemic toxicity exhibited by free HF while maintaining the tumor-suppressive properties of HF. LC-MS/MS analysis revealed that the reduction in the magnitude of adverse effects was the result of the ability to release HF from the HFm core in a slow and sustained manner. These results thus support the contention that HFm will potentially counteract Nrf2-activated cancers in the clinical settings.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Nanopartículas , Piperidinas , Quinazolinonas , Humanos , Adenocarcinoma del Pulmón/metabolismo , Cromatografía Liquida , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Neoplasias Pulmonares/patología , Micelas , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Piperidinas/farmacología , Quinazolinonas/farmacología , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA