Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Sci ; 115(5): 1576-1586, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38468443

RESUMEN

While loss of function (LOF) of retinoblastoma 1 (RB1) tumor suppressor is known to drive initiation of small-cell lung cancer and retinoblastoma, RB1 mutation is rarely observed in breast cancers at their initiation. In this study, we investigated the impact on untransformed mammary epithelial cells given by RB1 LOF. Depletion of RB1 in anon-tumorigenic MCF10A cells induced reversible growth arrest (quiescence) featured by downregulation of multiple cyclins and MYC, upregulation of p27KIP1, and lack of expression of markers which indicate cellular senescence or epithelial-mesenchymal transition (EMT). We observed a similar phenomenon in human mammary epithelial cells (HMEC) as well. Additionally, we found that RB1 depletion attenuated the activity of RAS and the downstream MAPK pathway in an RBL2/p130-dependent manner. The expression of farnesyltransferase ß, which is essential for RAS maturation, was found to be downregulated following RB1 depletion also in an RBL2/p130-dependent manner. These findings unveiled an unexpected mechanism whereby normal mammary epithelial cells resist to tumor initiation upon RB1 LOF.


Asunto(s)
Regulación hacia Abajo , Células Epiteliales , Proteínas de Unión a Retinoblastoma , Transducción de Señal , Proteínas ras , Humanos , Células Epiteliales/metabolismo , Femenino , Proteínas de Unión a Retinoblastoma/metabolismo , Proteínas de Unión a Retinoblastoma/genética , Proteínas ras/metabolismo , Proteínas ras/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Transición Epitelial-Mesenquimal/genética , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Glándulas Mamarias Humanas/citología , Línea Celular Tumoral , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética
2.
Cancer Sci ; 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38923741

RESUMEN

RECK has been described to modulate extracellular matrix components through negative regulation of MMP activities. Recently, RECK was demonstrated to bind to an orphan G protein-coupled receptor GPR124 to mediate WNT7 signaling in nontumor contexts. Here, we attempted to clarify the role of RECK in driving WNT signaling in cancer cells. RECK and GPR124 formed a complex in 293T cells, and when both were expressed, WNT signaling was significantly enhanced in a WNT7-dependent manner. This cooperation was abolished when RECK mutants unable to bind to GPR124 were transduced. RECK stimulated the growth of KRAS-mutated pancreatic ductal adenocarcinoma (PDAC) cells with increased sensitivity to WNT inhibitor in a GPR124-dependent manner. A gastric cancer cell line SH10TC endogenously expresses both RECK and GPR124 under regular culture conditions. In this cell line, inhibited cell growth and WNT signaling as well as increased apoptosis in the GPR124 depletion was dominantly found over those in the RECK deletion. These findings suggest that RECK promotes tumor cell growth by positively modulating WNT signaling through GPR124. This study proposes that the RECK/GPR124 complex might be a good therapeutic target in PDAC and gastric cancer.

3.
Bioorg Med Chem ; 102: 117674, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38457912

RESUMEN

Controlling RAS mutant cancer progression remains a significant challenge in developing anticancer drugs. Whereas Ras G12C-covalent binders have received clinical approval, the emergence of further mutations, along with the activation of Ras-related proteins and signals, has led to resistance to Ras binders. To discover novel compounds to overcome this bottleneck, we focused on the concurrent and sustained blocking of two major signaling pathways downstream of Ras. To this end, we synthesized 25 drug-drug conjugates (DDCs) by combining the MEK inhibitor trametinib with Akt inhibitors using seven types of linkers with structural diversity. The DDCs were evaluated for their cell permeability/accumulation and ability to inhibit proliferation in RAS-mutant cell lines. A representative DDC was further evaluated for its effects on signaling proteins, induction of apoptosis-related proteins, and the stability of hepatic metabolic enzymes. These in vitro studies identified a series of DDCs, especially those containing a furan-based linker, with promising properties as agents for treating RAS-mutant cancers. Additionally, in vivo experiments in mice using the two selected DDCs revealed prolonged half-lives and anticancer efficacies comparable to those of trametinib. The PK profiles of trametinib and the Akt inhibitor were unified through the DDC formation. The DDCs developed in this study have potential as drug candidates for the broad inhibition of RAS-mutant cancers.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Transducción de Señal , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Mutación , Inhibidores de la Angiogénesis/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Línea Celular Tumoral
4.
Chemistry ; 29(69): e202302297, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-37743326

RESUMEN

Chemists have long sought to regulate the reactivity of H2 , to yield hydride ions, hydrogen atoms, or electrons on demand. One source of inspiration for achieving this control is [NiFe]hydrogenase ([NiFe]H2 ase), which reacts with H2 to form various hydrogen active species such as NiIII hydride species, NiII hydride species, and NiI low-valent species. Chemists have attempted to synthesize these hydrogen active species not only as models for the active species of [NiFe]H2 ase, but also as electron transfer catalysts. However, the synthesis of NiI complex directly from H2 has not been reported. This paper reports the first example of a single-step synthesis of a NiI complex, via reaction of a NiII complex with H2 , stable for over 3 months at room temperature and we further demonstrate a reductive coupling of acridinium ions as part of a reaction cycle.

5.
Biochem Biophys Res Commun ; 621: 74-79, 2022 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-35810594

RESUMEN

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of immature T lymphocytes. Although various therapeutic approaches have been developed, refractoriness of chemotherapy and relapse cause a poor prognosis of the disease and further therapeutic strategies are required. Here, we report that Ras homolog enriched in brain (RHEB), a critical regulator of mTOR complex 1 activity, is a potential target for T-ALL therapy. In this study, we established an sgRNA library that comprehensively targeted mTOR upstream and downstream pathways, including autophagy. CRISPR/Cas9 dropout screening revealed critical roles of mTOR-related molecules in T-ALL cell survival. Among the regulators, we focused on RHEB because we previously found that it is dispensable for normal hematopoiesis in mice. Transcriptome and metabolic analyses revealed that RHEB deficiency suppressed de novo nucleotide biosynthesis, leading to human T-ALL cell death. Importantly, RHEB deficiency suppressed tumor growth in both mouse and xenograft models. Our data provide a potential strategy for efficient therapy of T-ALL by RHEB-specific inhibition.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Proteína Homóloga de Ras Enriquecida en el Cerebro , Animales , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
6.
Hepatology ; 74(4): 1971-1993, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33931882

RESUMEN

BACKGROUND AND AIMS: Synthetic cyclin-dependent kinase (CDK) 4/6 inhibitors exert antitumor effects by forcing RB1 in unphosphorylated status, causing not only cell cycle arrest but also cellular senescence, apoptosis, and increased immunogenicity. These agents currently have an indication in advanced breast cancers and are in clinical trials for many other solid tumors. HCC is one of promising targets of CDK4/6 inhibitors. RB family dysfunction is often associated with the initiation of HCC; however, this is revivable, as RB family members are not frequently mutated or deleted in this malignancy. APPROACH AND RESULTS: Loss of all Rb family members in transformation related protein 53 (Trp53)-/- mouse liver resulted in liver tumor reminiscent of human HCC, and re-expression of RB1 sensitized these tumors to a CDK4/6 inhibitor, palbociclib. Introduction of an unphosphorylatable form of RB1 (RB7LP) into multiple liver tumor cell lines induced effects similar to palbociclib. By screening for compounds that enhance the efficacy of RB7LP, we identified an I kappa B kinase (IKK)ß inhibitor Bay 11-7082. Consistently, RB7LP expression and treatment with palbociclib enhanced IKKα/ß phosphorylation and NF-κB activation. Combination therapy using palbociclib with Bay 11-7082 was significantly more effective in hepatoblastoma and HCC treatment than single administration. Moreover, blockade of IKK-NF-κB or AKT pathway enhanced effects of palbociclib on RB1-intact KRAS Kirsten rat sarcoma viral oncogene homolog mutated lung and colon cancers. CONCLUSIONS: In conclusion, CDK4/6 inhibitors have a potential to treat a wide variety of RB1-intact cancers including HCC when combined with an appropriate kinase inhibitor.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Aminopiridinas/farmacología , Aminopiridinas/uso terapéutico , Animales , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Carcinoma Hepatocelular/genética , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Células Hep G2 , Humanos , Técnicas In Vitro , Neoplasias Hepáticas/genética , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/genética , Ratones , Trasplante de Neoplasias , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Purinas/farmacología , Purinas/uso terapéutico , Piridinas/uso terapéutico , Proteína de Retinoblastoma , Proteína p53 Supresora de Tumor/genética , Proteínas de Xenopus
7.
FASEB J ; 35(5): e21503, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811686

RESUMEN

The muscle regeneration process requires a properly assembled extracellular matrix (ECM). Its homeostasis depends on the activity of different matrix-metalloproteinases (MMPs). The reversion-inducing-cysteine-rich protein with kazal motifs (RECK) is a membrane-anchored protein that negatively regulates the activity of different MMPs. However, the role of RECK in the process of skeletal muscle differentiation, regeneration, and fibrosis has not been elucidated. Here, we show that during skeletal muscle differentiation of C2C12 myoblasts and in satellite cells on isolated muscle fibers, RECK is transiently up regulated. C2C12 myoblasts with reduced RECK levels are more prone to enter the differentiation program, showing an accelerated differentiation process. Notch-1 signaling was reduced, while p38 and AKT signaling were augmented in myoblasts with decreased RECK levels. Overexpression of RECK restores the normal differentiation process but diminished the ability to form myotubes. Transient up-regulation of RECK occurs during skeletal muscle regeneration, which was accelerated in RECK-deficient mice (Reck±). RECK, MMPs and ECM proteins augmented in chronically damaged WT muscle, a model of muscle fibrosis. In this model, RECK ± mice showed diminished fibrosis compared to WT. These results strongly suggest that RECK is acting as a potential myogenic repressor during muscle formation and regeneration, emerging as a new player in these processes, and as a potential target to treat individuals with the muscle-wasting disease.


Asunto(s)
Diferenciación Celular , Matriz Extracelular/metabolismo , Fibrosis/prevención & control , Proteínas Ligadas a GPI/antagonistas & inhibidores , Desarrollo de Músculos , Músculo Esquelético/citología , Regeneración , Animales , Fibrosis/metabolismo , Fibrosis/patología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Transducción de Señal
8.
Cancer Sci ; 111(7): 2336-2348, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32437590

RESUMEN

Dietary fat consumption during accelerated stages of mammary gland development, such as peripubertal maturation or pregnancy, is known to increase the risk for breast cancer. However, the underlying molecular mechanisms are not fully understood. Here we examined the gene expression profile of mouse mammary epithelial cells (MMECs) on exposure to a high-fat diet (HFD) or control diet (CD). Trp53-/- female mice were fed with the experimental diets for 5 weeks during the peripubertal period (3-8 weeks of age). The treatment showed no significant difference in body weight between the HFD-fed mice and CD-fed mice. However, gene set enrichment analysis predicted a significant enrichment of c-Myc target genes in animals fed HFD. Furthermore, we detected enhanced activity and stabilization of c-Myc protein in MMECs exposed to a HFD. This was accompanied by augmented c-Myc phosphorylation at S62 with a concomitant increase in ERK phosphorylation. Moreover, MMECs derived from HFD-fed Trp53-/- mouse showed increased colony- and sphere-forming potential that was dependent on c-Myc. Further, oleic acid, a major fatty acid constituent of the HFD, and TAK-875, an agonist to G protein-coupled receptor 40 (a receptor for oleic acid), enhanced c-Myc stabilization and MMEC proliferation. Overall, our data indicate that HFD influences MMECs by stabilizing an oncoprotein, pointing to a novel mechanism underlying dietary fat-mediated mammary carcinogenesis.


Asunto(s)
Dieta Alta en Grasa , Epitelio/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Maduración Sexual , Animales , Línea Celular Tumoral , Femenino , Genes p53 , Humanos , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Pubertad , Células Tumorales Cultivadas
9.
Int J Mol Sci ; 21(7)2020 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-32244804

RESUMEN

The RB gene is one of the most frequently mutated genes in human cancers. Canonically, RB exerts its tumor suppressive activity through the regulation of the G1/S transition during cell cycle progression by modulating the activity of E2F transcription factors. However, aberration of the RB gene is most commonly detected in tumors when they gain more aggressive phenotypes, including metastatic activity or drug resistance, rather than accelerated proliferation. This implicates RB controls' malignant progression to a considerable extent in a cell cycle-independent manner. In this review, we highlight the multifaceted functions of the RB protein in controlling tumor lineage plasticity, metabolism, and the tumor microenvironment (TME), with a focus on the mechanism whereby RB controls the TME. In brief, RB inactivation in several types of cancer cells enhances production of pro-inflammatory cytokines, including CCL2, through upregulation of mitochondrial reactive oxygen species (ROS) production. These factors not only accelerate the growth of cancer cells in a cell-autonomous manner, but also stimulate non-malignant cells in the TME to generate a pro-tumorigenic niche in a non-cell-autonomous manner. Here, we discuss the biological and pathological significance of the non-cell-autonomous functions of RB and attempt to predict their potential clinical relevance to cancer immunotherapy.


Asunto(s)
Ciclo Celular/genética , Neoplasias/genética , Proteína de Retinoblastoma/genética , Microambiente Tumoral/genética , Proteínas Supresoras de Tumor/genética , Citocinas/metabolismo , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas Supresoras de Tumor/metabolismo
10.
J Biol Chem ; 291(41): 21496-21509, 2016 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-27519418

RESUMEN

Although abnormal metabolic regulation is a critical determinant of cancer cell behavior, it is still unclear how an altered balance between ATP production and consumption contributes to malignancy. Here we show that disruption of this energy balance efficiently suppresses aggressive malignant gliomas driven by mammalian target of rapamycin complex 1 (mTORC1) hyperactivation. In a mouse glioma model, mTORC1 hyperactivation induced by conditional Tsc1 deletion increased numbers of glioma-initiating cells (GICs) in vitro and in vivo Metabolic analysis revealed that mTORC1 hyperactivation enhanced mitochondrial biogenesis, as evidenced by elevations in oxygen consumption rate and ATP production. Inhibition of mitochondrial ATP synthetase was more effective in repressing sphere formation by Tsc1-deficient glioma cells than that by Tsc1-competent glioma cells, indicating a crucial function for mitochondrial bioenergetic capacity in GIC expansion. To translate this observation into the development of novel therapeutics targeting malignant gliomas, we screened drug libraries for small molecule compounds showing greater efficacy in inhibiting the proliferation/survival of Tsc1-deficient cells compared with controls. We identified several compounds able to preferentially inhibit mitochondrial activity, dramatically reducing ATP levels and blocking glioma sphere formation. In human patient-derived glioma cells, nigericin, which reportedly suppresses cancer stem cell properties, induced AMPK phosphorylation that was associated with mTORC1 inactivation and induction of autophagy and led to a marked decrease in sphere formation with loss of GIC marker expression. Furthermore, malignant characteristics of human glioma cells were markedly suppressed by nigericin treatment in vivo Thus, targeting mTORC1-driven processes, particularly those involved in maintaining a cancer cell's energy balance, may be an effective therapeutic strategy for glioma patients.


Asunto(s)
Metabolismo Energético , Glioma/metabolismo , Glioma/terapia , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Glioma/genética , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , ATPasas de Translocación de Protón Mitocondriales/genética , Complejos Multiproteicos/genética , Serina-Treonina Quinasas TOR/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
11.
Cancer Sci ; 108(9): 1726-1731, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28865172

RESUMEN

The Retinoblastoma (RB) tumor suppressor regulates G1 /S transition during cell cycle progression by modulating the activity of E2F transcription factors. The RB pathway plays a central role in the suppression of most cancers, and RB mutation was initially discovered by virtue of its role in tumor initiation. However, as cancer genome sequencing has evolved to profile more advanced and treatment-resistant cancers, it has become increasingly clear that, in the majority of cancers, somatic RB inactivation occurs during tumor progression. Furthermore, despite the presence of deregulation of cell cycle control due to an INK4A deletion, additional CCND amplification and/or other mutations in the RB pathway, mutation or deletion of the RB gene is often observed during cancer progression. Of note, RB inactivation during cancer progression not only facilitates G1 /S transition but also enhances some characteristics of malignancy, including altered drug sensitivity and a return to the undifferentiated state. Recently, we reported that RB inactivation enhances pro-inflammatory signaling through stimulation of the interleukin-6/STAT3 pathway, which directly promotes various malignant features of cancer cells. In this review, we highlight the consequences of RB inactivation during cancer progression, and discuss the biological and pathological significance of the interaction between RB and pro-inflammatory signaling.


Asunto(s)
Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteína de Retinoblastoma/fisiología , Animales , Transformación Celular Neoplásica/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Metabolismo Energético , Genes Supresores de Tumor , Humanos , Inflamación/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Transducción de Señal
12.
J Pediatr Hematol Oncol ; 39(5): e285-e289, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28267084

RESUMEN

Liver fibrosis is one of the common complications of transient myeloproliferative disorder (TMD) in Down syndrome (DS), but the exact molecular pathogenesis is largely unknown. We herein report a neonate of DS with liver fibrosis associated with TMD, in which we performed the serial profibrogenic cytokines analyses. We found the active monocyte chemoattractant protein-1 expression in the affected liver tissue and also found that both serum and urinary monocyte chemoattractant protein-1 concentrations are noninvasive biomarkers of liver fibrosis. We also showed a prospective of the future anticytokine therapy with herbal medicine for the liver fibrosis associated with TMD in DS.


Asunto(s)
Quimiocina CCL2/análisis , Síndrome de Down/complicaciones , Reacción Leucemoide/complicaciones , Cirrosis Hepática/diagnóstico , Biomarcadores , Citocinas/análisis , Diagnóstico Diferencial , Humanos , Recién Nacido , Hígado/química , Hígado/patología , Cirrosis Hepática/etiología
13.
Mol Carcinog ; 55(12): 1974-1989, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26621780

RESUMEN

Mutations in RB and PTEN are linked to castration resistance and poor prognosis in prostate cancer. Identification of genes that are regulated by these tumor suppressors in a context that recapitulates cancer progression may be beneficial for discovering novel therapeutic targets. Although various genetically engineered mice thus far provided tumor models with various pathological stages, they are not ideal for detecting dynamic changes in gene transcription. Additionally, it is difficult to achieve an effect specific to tumor progression via gain of functions of these genes. In this study, we developed an in vitro model to help identify RB- and PTEN-loss signatures during the malignant progression of prostate cancers. Trp53-/- ; Rbf/f , Trp53-/- ; Ptenf/f , and Trp53-/- ; Rbf/f ; Ptenf/f prostate epithelial cells were infected with AD-LacZ or AD-Cre. We found that deletion of Rb, Pten or both stimulated prostasphere formation and tumor development in immune-compromised mice. The GO analysis of genes affected by the deletion of Rb or Pten in Trp53-/- prostate epithelial cells identified a number of genes encoding cytokines, chemokines and extracellular matrix remodeling factors, but only few genes related to cell cycle progression. Two genes (Il-6 and Lox) were further analyzed. Blockade of Il-6 signaling and depletion of Lox significantly attenuated prostasphere formation in 3D culture, and in the case of IL-6, strongly suppressed tumor growth in vivo. These findings suggest that our in vitro model may be instrumental in identifying novel therapeutic targets of prostate cancer progression, and further underscore IL-6 and LOX as promising therapeutic targets. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Carcinogénesis/patología , Regulación Neoplásica de la Expresión Génica , Fosfohidrolasa PTEN/genética , Próstata/patología , Neoplasias de la Próstata/patología , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis/genética , Células Cultivadas , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Eliminación de Gen , Masculino , Ratones , Ratones Noqueados , Próstata/metabolismo , Neoplasias de la Próstata/genética , Transducción de Señal
14.
Stem Cells ; 33(5): 1657-69, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25694388

RESUMEN

Retinoblastoma tumor suppressor protein (RB) is inactivated more frequently during tumor progression than during tumor initiation. However, its exact role in controlling the malignant features associated with tumor progression is poorly understood. We established in vivo and in vitro models to investigate the undifferentiated state induced by Rb inactivation. Rb heterozygous mice develop well-differentiated thyroid medullary carcinoma. We found that additional deletion of Trp53, without change in lineage, converted these Rb-deficient tumors to a poorly differentiated type associated with higher self-renewal activity. Freshly prepared mouse embryonic fibroblasts (MEFs) of Rb(-/-) ; Trp53(-/-) background formed stem cell-like spheres that expressed significant levels of embryonic genes despite of lacking the ability to form colonies on soft agar or tumors in immune-deficient mice. This suggested that Rb-p53 double inactivation resulted in an undifferentiated status but without carcinogenic conversion. We next established Rb(-/-) ; N-ras(-/-) MEFs that harbored a spontaneous carcinogenic mutation in Trp53. These cells (RN6), in an Rb-dependent manner, efficiently generated spheres that expressed very high levels of embryonic genes, and appeared to be carcinogenic. We then screened an FDA-approved drug library to search for agents that suppressed the spherogenic activity of RN6 cells. Data revealed that RN6 cells were sensitive to specific agents including ones those are effective against cancer stem cells. Taken together, all these findings suggest that the genetic interaction between Rb and p53 is a critical determinant of the undifferentiated state in normal and tumor cells.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/citología , Fibroblastos/citología , Células Neuroendocrinas/citología , Proteína de Retinoblastoma/metabolismo , Glándula Tiroides/citología , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Conducta Animal , Línea Celular , Evaluación Preclínica de Medicamentos , Fibroblastos/metabolismo , Heterocigoto , Ratones Noqueados , Datos de Secuencia Molecular , Mutación/genética , Fenotipo , Proteína de Retinoblastoma/deficiencia , Esferoides Celulares/metabolismo , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteínas ras/metabolismo
15.
J Biol Chem ; 289(34): 23417-27, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25006242

RESUMEN

The acquisition of skeletal muscle-specific function and terminal cell cycle arrest represent two important features of the myogenic differentiation program. These cellular processes are distinct and can be separated genetically. The lineage-specific transcription factor MyoD and the retinoblastoma protein pRb participate in both of these cellular events. Whether and how MyoD and pRb work together to effect terminal cell cycle arrest is uncertain. To address this question, we focused on cyclin D1, whose stable repression is required for terminal cell cycle arrest and execution of myogenesis. MyoD and pRb are both required for the repression of cyclin D1; their actions, however, were found not to be direct. Rather, they operate to regulate the immediate early gene Fra-1, a critical player in mitogen-dependent induction of cyclin D1. Two conserved MyoD-binding sites were identified in an intronic enhancer of Fra-1 and shown to be required for the stable repression of Fra-1 and, in turn, cyclin D1. Localization of MyoD alone to the intronic enhancer of Fra-1 in the absence of pRb was not sufficient to elicit a block to Fra-1 induction; pRb was also recruited to the intronic enhancer in a MyoD-dependent manner. These observations suggest that MyoD and pRb work together cooperatively at the level of the intronic enhancer of Fra-1 during terminal cell cycle arrest. This work reveals a previously unappreciated link between a lineage-specific transcription factor, a tumor suppressor, and a proto-oncogene in the control of an important facet of myogenic differentiation.


Asunto(s)
Ciclo Celular , Ciclina D1/metabolismo , Desarrollo de Músculos , Proteína MioD/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteína de Retinoblastoma/fisiología , Células 3T3 , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Línea Celular Transformada , ADN , Cartilla de ADN , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Polimerasa II/metabolismo
16.
Nat Genet ; 38(1): 118-23, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16369533

RESUMEN

Mutations in the gene encoding the retinoblastoma tumor suppressor predispose humans and mice to tumor development. Here we have assessed the effect of Nras loss on tumor development in Rb1 heterozygous mice. Loss of one or two Nras alleles is shown to significantly reduce the severity of pituitary tumors arising in Rb1(+/-) animals by enhancing their differentiation. By contrast, C-cell thyroid adenomas occurring in Rb1(+/-) mice progress to metastatic medullary carcinomas after loss of Nras. In Rb1(+/-)Nras(+/-) animals, distant medullary thyroid carcinoma metastases are associated with loss of the remaining wild-type Nras allele. Loss of Nras in Rb1-deficient C cells results in elevated Ras homolog family A (RhoA) activity, and this is causally linked to the invasiveness and metastatic behavior of these cells. These findings suggest that the loss of the proto-oncogene Nras in certain cellular contexts can promote malignant tumor progression.


Asunto(s)
Genes ras/genética , Tumores Neuroendocrinos/patología , Proteína de Retinoblastoma/deficiencia , Neoplasias de la Tiroides/patología , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenoma/genética , Adenoma/patología , Hormona Adrenocorticotrópica/metabolismo , Amidas/farmacología , Animales , Carcinoma Medular/genética , Carcinoma Medular/patología , Inhibidores Enzimáticos/farmacología , Heterocigoto , Ratones , Ratones Mutantes , Ratones Desnudos , Metástasis de la Neoplasia , Tumores Neuroendocrinos/genética , Proto-Oncogenes Mas , Piridinas/farmacología , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/genética , Transducción de Señal , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/secundario , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
17.
Biochem Biophys Res Commun ; 450(1): 837-43, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24960197

RESUMEN

Acute myeloid leukaemia (AML) is a heterogeneous neoplastic disorder in which a subset of cells function as leukaemia-initiating cells (LICs). In this study, we prospectively evaluated the leukaemia-initiating capacity of AML cells fractionated according to the expression of a nucleolar GTP binding protein, nucleostemin (NS). To monitor NS expression in living AML cells, we generated a mouse AML model in which green fluorescent protein (GFP) is expressed under the control of a region of the NS promoter (NS-GFP). In AML cells, NS-GFP levels were correlated with endogenous NS mRNA. AML cells with the highest expression of NS-GFP were very immature blast-like cells, efficiently formed leukaemia colonies in vitro, and exhibited the highest leukaemia-initiating capacity in vivo. Gene expression profiling analysis revealed that cell cycle regulators and nucleotide metabolism-related genes were highly enriched in a gene set associated with leukaemia-initiating capacity that we termed the 'leukaemia stem cell gene signature'. This gene signature stratified human AML patients into distinct clusters that reflected prognosis, demonstrating that the mouse leukaemia stem cell gene signature is significantly associated with the malignant properties of human AML. Further analyses of gene regulation in leukaemia stem cells could provide novel insights into diagnostic and therapeutic approaches to AML.


Asunto(s)
Proteínas de Unión al GTP/genética , Predisposición Genética a la Enfermedad/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , Animales , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Marcadores Genéticos/genética , Humanos , Ratones , Ratones Transgénicos , Pronóstico
18.
JACS Au ; 4(4): 1615-1622, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38665645

RESUMEN

Have you ever imagined reactions of alkenes with hydrogen that result in anything other than hydrogenation or hydrogenative C-C coupling? We have long sought to develop not only hydrogenation catalysts that activate H2 as hydride ions but also electron transfer catalysts that activate H2 as a direct electron donor. Here, we report the reductive cyclopropanation of alkenes using an iridium electron storage catalyst with H2 as the electron source without releasing metal waste from the reductant. We discuss the catalytic mechanism with selectivity to give the trans-isomer. These findings are based on the isolation of three complexes and density functional theory calculations.

19.
Med Mycol J ; 65(1): 1-5, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38417881

RESUMEN

Microsporum canis is a type of dermatophyte that causes zoonotic dermatophytosis in cats and dogs. We report three cases of tinea corporis due to M. canis from a single household with a domestic cat as a pet. The cases included a woman in her thirties (mother), a girl in her teens (older sister), and a girl in her teens (younger sister). Following sudden hair loss in the domestic cat, annular erythema with pruritus and scales appeared on the face, neck, and limbs of the older sister, younger sister, and mother, sequentially; they subsequently visited our hospital. Potassium hydroxide direct microscopy revealed filamentous fungi on all three women. In addition, short-haired colonies with a white to yellowish-white color and extending in a radial manner were found in cultures using a flat plate agar medium. A slide culture with the same medium indicated pointed spindle-shaped macroconidia with 7-8 septa. Therefore, the cases were diagnosed as tinea corporis due to M. canis. Genetic analysis of the cells of the cat and the mother, older sister, and younger sister using multilocus microsatellite typing (MLMT) indicated that all cases were classified into the same genotype, suggesting that the transmission route of these cases was familial. Here, we show that MLMT is useful in identifying the infection route in cases of tinea corporis due to M. canis.


Asunto(s)
Dermatomicosis , Tiña , Humanos , Adolescente , Femenino , Animales , Perros , Gatos , Tiña/diagnóstico , Tiña/veterinaria , Microsporum/genética , Madres , Repeticiones de Microsatélite/genética , Dermatomicosis/diagnóstico , Dermatomicosis/microbiología
20.
Commun Biol ; 7(1): 740, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890503

RESUMEN

Although low estrogen is considered to suppress uterine endometrial carcinoma, the most cases occur in the postmenopausal stage. After menopause, the production of androgen level also declines. Therefore, to resolve the above enigma, we hypothesize that the postmenopausal decline of androgen is a trigger of its progression. In the present study, to validate this hypothesis, we examine the pathological roles of androgen/AR by analyzing clinical data, culturing endometrioid cancer cell lines, and using murine models. Clinical data show that androgen receptor (AR) expression and serum dihydrotestosterone (DHT) are associated with lower disease-free survival (DFS). DHT suppresses malignant behaviors in AR-transfected human endometrial cancer cells (ECC). In ovariectomized Ptenff/PRcre/+ mice, DHT decreases the proliferation of spontaneously developed murine ECC. In AR-transfected human ECC and Ptenff/PRcre/+ mice, DHT suppresses FOXP4 expression. FOXP4-overexpressed human ECC increases, while FOXP4-knocked-down ECC shows decreased malignant behaviors. DHT/AR-mediated ECC suppression is restored by FOXP4 overexpression. The high FOXP4 expression is significantly correlated with low postoperative DFS. These findings indicate that the androgen/AR system suppresses the malignant activity of endometrial carcinoma and that downstream FOXP4 is another target molecule. These findings will also impact developments in clinical approaches to elderly health.


Asunto(s)
Andrógenos , Neoplasias Endometriales , Factores de Transcripción Forkhead , Receptores Androgénicos , Femenino , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/genética , Humanos , Animales , Ratones , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Andrógenos/metabolismo , Línea Celular Tumoral , Dihidrotestosterona/metabolismo , Dihidrotestosterona/farmacología , Regulación Neoplásica de la Expresión Génica , Persona de Mediana Edad , Proliferación Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA