Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Gastroenterology ; 160(6): 2103-2118, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33465371

RESUMEN

BACKGROUND & AIMS: Liver tight junctions (TJs) establish tissue barriers that isolate bile from the blood circulation. TJP2/ZO-2-inactivating mutations cause progressive cholestatic liver disease in humans. Because the underlying mechanisms remain elusive, we characterized mice with liver-specific inactivation of Tjp2. METHODS: Tjp2 was deleted in hepatocytes, cholangiocytes, or both. Effects on the liver were assessed by biochemical analyses of plasma, liver, and bile and by electron microscopy, histology, and immunostaining. TJ barrier permeability was evaluated using fluorescein isothiocyanate-dextran (4 kDa). Cholic acid (CA) diet was used to assess susceptibility to liver injury. RESULTS: Liver-specific deletion of Tjp2 resulted in lower Cldn1 protein levels, minor changes to the TJ, dilated canaliculi, lower microvilli density, and aberrant radixin and bile salt export pump (BSEP) distribution, without an overt increase in TJ permeability. Hepatic Tjp2-defcient mice presented with mild progressive cholestasis with lower expression levels of bile acid transporter Abcb11/Bsep and detoxification enzyme Cyp2b10. A CA diet tolerated by control mice caused severe cholestasis and liver necrosis in Tjp2-deficient animals. 1,4-Bis[2-(3,5-dichloropyridyloxy)]benzene ameliorated CA-induced injury by enhancing Cyp2b10 expression, and ursodeoxycholic acid provided partial improvement. Inactivating Tjp2 separately in hepatocytes or cholangiocytes showed only mild CA-induced liver injury. CONCLUSION: Tjp2 is required for normal cortical distribution of radixin, canalicular volume regulation, and microvilli density. Its inactivation deregulated expression of Cldn1 and key bile acid transporters and detoxification enzymes. The mice provide a novel animal model for cholestatic liver disease caused by TJP2-inactivating mutations in humans.


Asunto(s)
Canalículos Biliares/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Colestasis/genética , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-2/genética , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/genética , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Ácidos y Sales Biliares/metabolismo , Canalículos Biliares/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Colagogos y Coleréticos/uso terapéutico , Ácido Cólico , Claudina-1/metabolismo , Familia 2 del Citocromo P450/metabolismo , Proteínas del Citoesqueleto/metabolismo , Células Epiteliales , Femenino , Fibrosis , Predisposición Genética a la Enfermedad , Hepatocitos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Mutación , Oxazoles/uso terapéutico , Permeabilidad , Factores Protectores , ARN Mensajero/metabolismo , Esteroide Hidroxilasas/metabolismo , Uniones Estrechas/ultraestructura , Ácido Ursodesoxicólico/uso terapéutico , Proteína de la Zonula Occludens-2/deficiencia
2.
Cell Mol Life Sci ; 77(17): 3423-3439, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31722069

RESUMEN

SUGCT (C7orf10) is a mitochondrial enzyme that synthesizes glutaryl-CoA from glutarate in tryptophan and lysine catabolism, but it has not been studied in vivo. Although mutations in Sugct lead to Glutaric Aciduria Type 3 disease in humans, patients remain largely asymptomatic despite high levels of glutarate in the urine. To study the disease mechanism, we generated SugctKO mice and uncovered imbalanced lipid and acylcarnitine metabolism in kidney in addition to changes in the gut microbiome. After SugctKO mice were treated with antibiotics, metabolites were comparable to WT, indicating that the microbiome affects metabolism in SugctKO mice. SUGCT loss of function contributes to gut microbiota dysbiosis, leading to age-dependent pathological changes in kidney, liver, and adipose tissue. This is associated with an obesity-related phenotype that is accompanied by lipid accumulation in kidney and liver, as well as "crown-like" structures in adipocytes. Furthermore, we show that the SugctKO kidney pathology is accelerated and exacerbated by a high-lysine diet. Our study highlights the importance of non-essential genes with no readily detectable early phenotype, but with substantial contributions to the development of age-related pathologies, which result from an interplay between genetic background, microbiome, and diet in the health of mammals.


Asunto(s)
Envejecimiento , Coenzima A Transferasas/genética , Microbioma Gastrointestinal , Síndrome Metabólico/patología , Animales , Antibacterianos/farmacología , Bacterias/genética , Bacterias/aislamiento & purificación , Carnitina/análogos & derivados , Carnitina/metabolismo , Coenzima A Transferasas/deficiencia , Suplementos Dietéticos , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Riñón/metabolismo , Riñón/patología , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología , Lisina/administración & dosificación , Síndrome Metabólico/metabolismo , Metaboloma/efectos de los fármacos , Ratones , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Triptófano/metabolismo
3.
Am J Hum Genet ; 101(3): 391-403, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886341

RESUMEN

In five separate families, we identified nine individuals affected by a previously unidentified syndrome characterized by growth retardation, spine malformation, facial dysmorphisms, and developmental delays. Using homozygosity mapping, array CGH, and exome sequencing, we uncovered bi-allelic loss-of-function CDK10 mutations segregating with this disease. CDK10 is a protein kinase that partners with cyclin M to phosphorylate substrates such as ETS2 and PKN2 in order to modulate cellular growth. To validate and model the pathogenicity of these CDK10 germline mutations, we generated conditional-knockout mice. Homozygous Cdk10-knockout mice died postnatally with severe growth retardation, skeletal defects, and kidney and lung abnormalities, symptoms that partly resemble the disease's effect in humans. Fibroblasts derived from affected individuals and Cdk10-knockout mouse embryonic fibroblasts (MEFs) proliferated normally; however, Cdk10-knockout MEFs developed longer cilia. Comparative transcriptomic analysis of mutant and wild-type mouse organs revealed lipid metabolic changes consistent with growth impairment and altered ciliogenesis in the absence of CDK10. Our results document the CDK10 loss-of-function phenotype and point to a function for CDK10 in transducing signals received at the primary cilia to sustain embryonic and postnatal development.


Asunto(s)
Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/fisiología , Discapacidades del Desarrollo/genética , Trastornos del Crecimiento/genética , Mutación , Columna Vertebral/anomalías , Columna Vertebral/patología , Animales , Ciclo Celular , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Cilios/metabolismo , Cilios/patología , Discapacidades del Desarrollo/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Trastornos del Crecimiento/patología , Humanos , Lactante , Masculino , Ratones , Ratones Noqueados , Linaje , Fosforilación , Transducción de Señal , Columna Vertebral/metabolismo
4.
Allergy ; 74(9): 1660-1674, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30891811

RESUMEN

Innate immune system quickly responds to invasion of microbes and foreign substances through the extracellular and intracellular sensing receptors, which recognize distinctive molecular and structural patterns. The recognition of innate immune receptors leads to the induction of inflammatory and adaptive immune responses by activating downstream signaling pathways. Allergy is an immune-related disease and results from a hypersensitive immune response to harmless substances in the environment. However, less is known about the activation of innate immunity during exposure to allergens. New insights into the innate immune system by sensors and their signaling cascades provide us with more important clues and a framework for understanding allergy disorders. In this review, we will focus on recent advances in the innate immune sensing system.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Hipersensibilidad/etiología , Inmunidad Innata , Inmunidad Adaptativa , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Humanos , Hipersensibilidad/diagnóstico , Hipersensibilidad/metabolismo , Inflamasomas , Lectinas Tipo C/metabolismo , FN-kappa B/metabolismo , Proteínas NLR/metabolismo , Proteínas Quinasas/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Células Th2/inmunología , Células Th2/metabolismo , Receptores Toll-Like/metabolismo
5.
Bioessays ; 39(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28752618

RESUMEN

The maintenance of cell size homeostasis has been studied for years in different cellular systems. With the focus on 'what regulates cell size', the question 'why cell size needs to be maintained' has been largely overlooked. Recent evidence indicates that animal cells exhibit nonlinear cell size dependent growth rates and mitochondrial metabolism, which are maximal in intermediate sized cells within each cell population. Increases in intracellular distances and changes in the relative cell surface area impose biophysical limitations on cells, which can explain why growth and metabolic rates are maximal in a specific cell size range. Consistently, aberrant increases in cell size, for example through polyploidy, are typically disadvantageous to cellular metabolism, fitness and functionality. Accordingly, cellular hypertrophy can potentially predispose to or worsen metabolic diseases. We propose that cell size control may have emerged as a guardian of cellular fitness and metabolic activity.


Asunto(s)
Homeostasis/fisiología , Animales , Tamaño de la Célula , Humanos , Hipertrofia/fisiopatología , Mitocondrias/metabolismo , Mitocondrias/fisiología
6.
PLoS Genet ; 12(9): e1006310, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27631493

RESUMEN

The Greatwall kinase/Mastl is an essential gene that indirectly inhibits the phosphatase activity toward mitotic Cdk1 substrates. Here we show that although Mastl knockout (MastlNULL) MEFs enter mitosis, they progress through mitosis without completing cytokinesis despite the presence of misaligned chromosomes, which causes chromosome segregation defects. Furthermore, we uncover the requirement of Mastl for robust spindle assembly checkpoint (SAC) maintenance since the duration of mitotic arrest caused by microtubule poisons in MastlNULL MEFs is shortened, which correlates with premature disappearance of the essential SAC protein Mad1 at the kinetochores. Notably, MastlNULL MEFs display reduced phosphorylation of a number of proteins in mitosis, which include the essential SAC kinase MPS1. We further demonstrate that Mastl is required for multi-site phosphorylation of MPS1 as well as robust MPS1 kinase activity in mitosis. In contrast, treatment of MastlNULL cells with the phosphatase inhibitor okadaic acid (OKA) rescues the defects in MPS1 kinase activity, mislocalization of phospho-MPS1 as well as Mad1 at the kinetochore, and premature SAC silencing. Moreover, using in vitro dephosphorylation assays, we demonstrate that Mastl promotes persistent MPS1 phosphorylation by inhibiting PP2A/B55-mediated MPS1 dephosphorylation rather than affecting Cdk1 kinase activity. Our findings establish a key regulatory function of the Greatwall kinase/Mastl->PP2A/B55 pathway in preventing premature SAC silencing.


Asunto(s)
Proteína Quinasa CDC2/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Proteínas Asociadas a Microtúbulos/genética , Proteína Fosfatasa 2/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Segregación Cromosómica/genética , Citocinesis/genética , Cinetocoros/metabolismo , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/genética , Microtúbulos/metabolismo , Mitosis/genética , Fosforilación , Proteína Fosfatasa 2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/genética
7.
Haematologica ; 100(4): 431-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25616574

RESUMEN

Mouse knockouts of Cdk2 and Cdk4 are individually viable whereas the double knockouts are embryonic lethal due to heart defects, and this precludes the investigation of their overlapping roles in definitive hematopoiesis. Here we use a conditional knockout mouse model to investigate the effect of combined loss of Cdk2 and Cdk4 in hematopoietic cells. Cdk2(fl/fl)Cdk4(-/-)vavCre mice are viable but displayed a significant increase in erythrocyte size. Cdk2(fl/fl)Cdk4(-/-)vavCre mouse bone marrow exhibited reduced phosphorylation of the retinoblastoma protein and reduced expression of E2F target genes such as cyclin A2 and Cdk1. Erythroblasts lacking Cdk2 and Cdk4 displayed a lengthened G1 phase due to impaired phosphorylation of the retinoblastoma protein. Deletion of the retinoblastoma protein rescued the increased size displayed by erythrocytes lacking Cdk2 and Cdk4, indicating that the retinoblastoma/Cdk2/Cdk4 pathway regulates erythrocyte size. The recovery of platelet counts following a 5-fluorouracil challenge was delayed in Cdk2(fl/fl)Cdk4(-/-)vavCre mice revealing a critical role for Cdk2 and Cdk4 in stress hematopoiesis. Our data indicate that Cdk2 and Cdk4 play important overlapping roles in homeostatic and stress hematopoiesis, which need to be considered when using broad-spectrum cyclin-dependent kinase inhibitors for cancer therapy.


Asunto(s)
Plaquetas/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Eritrocitos/citología , Hematopoyesis/genética , Estrés Fisiológico , Animales , Tamaño de la Célula , Quinasa 2 Dependiente de la Ciclina/deficiencia , Quinasa 4 Dependiente de la Ciclina/deficiencia , Quinasa 4 Dependiente de la Ciclina/genética , Femenino , Eliminación de Gen , Hematócrito , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Masculino , Ratones , Ratones Noqueados , Fenotipo , Ploidias , Proteína de Retinoblastoma/genética
8.
Oncogene ; 40(13): 2339-2354, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33664451

RESUMEN

Most of the drugs currently prescribed for cancer treatment are riddled with substantial side effects. In order to develop more effective and specific strategies to treat cancer, it is of importance to understand the biology of drug targets, particularly the newly emerging ones. A comprehensive evaluation of these targets will benefit drug development with increased likelihood for success in clinical trials. The folate-mediated one-carbon (1C) metabolism pathway has drawn renewed attention as it is often hyperactivated in cancer and inhibition of this pathway displays promise in developing anticancer treatment with fewer side effects. Here, we systematically review individual enzymes in the 1C pathway and their compartmentalization to mitochondria and cytosol. Based on these insight, we conclude that (1) except the known 1C targets (DHFR, GART, and TYMS), MTHFD2 emerges as good drug target, especially for treating hematopoietic cancers such as CLL, AML, and T-cell lymphoma; (2) SHMT2 and MTHFD1L are potential drug targets; and (3) MTHFD2L and ALDH1L2 should not be considered as drug targets. We highlight MTHFD2 as an excellent therapeutic target and SHMT2 as a complementary target based on structural/biochemical considerations and up-to-date inhibitor development, which underscores the perspectives of their therapeutic potential.


Asunto(s)
Linfoma de Células T/tratamiento farmacológico , Redes y Vías Metabólicas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Carbono/metabolismo , Ácido Fólico/genética , Ácido Fólico/metabolismo , Humanos , Linfoma de Células T/metabolismo , Linfoma de Células T/patología , Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias/metabolismo
9.
Hepatol Int ; 14(4): 463-474, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32578019

RESUMEN

Cell division is essential for organismal growth and tissue homeostasis. It is exceptionally significant in tissues chronically exposed to intrinsic and external damage, like the liver. After decades of studying the regulation of cell cycle by extracellular signals, there are still gaps in our knowledge on how these two interact with metabolic pathways in vivo. Studying the cross-talk of these pathways has direct clinical implications as defects in cell division, signaling pathways, and metabolic homeostasis are frequently observed in liver diseases. In this review, we will focus on recent reports which describe various functions of cell cycle regulators in hepatic homeostasis. We will describe the interplay between the cell cycle and metabolism during liver regeneration after acute and chronic damage. We will focus our attention on non-alcoholic fatty liver disease, especially non-alcoholic steatohepatitis. The global incidence of non-alcoholic fatty liver disease is increasing exponentially. Therefore, understanding the interplay between cell cycle regulators and metabolism may lead to the discovery of novel therapeutic targets amenable to intervention.


Asunto(s)
Ciclo Celular , Homeostasis , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Humanos , Enfermedad del Hígado Graso no Alcohólico/patología
10.
Elife ; 92020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33345777

RESUMEN

Cell cycle progression and lipid metabolism are well-coordinated processes required for proper cell proliferation. In liver diseases that arise from dysregulated lipid metabolism, proliferation is diminished. To study the outcome of CDK1 loss and blocked hepatocyte proliferation on lipid metabolism and the consequent impact on whole-body physiology, we performed lipidomics, metabolomics, and RNA-seq analyses on a mouse model. We observed reduced triacylglycerides in liver of young mice, caused by oxidative stress that activated FOXO1 to promote expression of Pnpla2/ATGL. Additionally, we discovered that hepatocytes displayed malfunctioning ß-oxidation, reflected by increased acylcarnitines (ACs) and reduced ß-hydroxybutyrate. This led to elevated plasma free fatty acids (FFAs), which were transported to the adipose tissue for storage and triggered greater insulin secretion. Upon aging, chronic hyperinsulinemia resulted in insulin resistance and hepatic steatosis through activation of LXR. Here, we demonstrate that loss of hepatocyte proliferation is not only an outcome but also possibly a causative factor for liver pathology.


Asunto(s)
Proteína Quinasa CDC2/fisiología , División Celular/fisiología , Hepatocitos/fisiología , Hiperinsulinismo/metabolismo , Metabolismo de los Lípidos , Tejido Adiposo/química , Tejido Adiposo/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Ácidos Grasos no Esterificados/sangre , Ácidos Grasos no Esterificados/metabolismo , Hepatocitos/metabolismo , Resistencia a la Insulina , Hígado/química , Hígado/metabolismo , Hígado/fisiología , Hepatopatías/etiología , Hepatopatías/metabolismo , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo
11.
Nat Commun ; 11(1): 3520, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32665551

RESUMEN

PRDM (PRDI-BF1 and RIZ homology domain containing) family members are sequence-specific transcriptional regulators involved in cell identity and fate determination, often dysregulated in cancer. The PRDM15 gene is of particular interest, given its low expression in adult tissues and its overexpression in B-cell lymphomas. Despite its well characterized role in stem cell biology and during early development, the role of PRDM15 in cancer remains obscure. Herein, we demonstrate that while PRDM15 is largely dispensable for mouse adult somatic cell homeostasis in vivo, it plays a critical role in B-cell lymphomagenesis. Mechanistically, PRDM15 regulates a transcriptional program that sustains the activity of the PI3K/AKT/mTOR pathway and glycolysis in B-cell lymphomas. Abrogation of PRDM15 induces a metabolic crisis and selective death of lymphoma cells. Collectively, our data demonstrate that PRDM15 fuels the metabolic requirement of B-cell lymphomas and validate it as an attractive and previously unrecognized target in oncology.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Inmunoprecipitación de Cromatina , Biología Computacional , Proteínas de Unión al ADN/genética , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Humanos , Linfoma/genética , Linfoma/metabolismo , Ratones , Ratones SCID , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Distribución Aleatoria , Factores de Transcripción/genética , Transcriptoma/genética
12.
Dev Cell ; 47(4): 425-438.e5, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30344111

RESUMEN

Liver disease is linked to a decreased capacity of hepatocytes to divide. In addition, cellular metabolism is important for tissue homeostasis and regeneration. Since metabolic changes are a hallmark of liver disease, we investigated the connections between metabolism and cell division. We determined global metabolic changes at different stages of liver regeneration using a combination of integrated transcriptomic and metabolomic analyses with advanced functional redox in vivo imaging. Our data indicate that blocking hepatocyte division during regeneration leads to mitochondrial dysfunction and downregulation of oxidative pathways. This resulted in an increased redox ratio and hyperactivity of alanine transaminase allowing the production of alanine and α-ketoglutarate from pyruvate when mitochondrial functions are impaired. Our data suggests that during liver regeneration, cell division leads to hepatic metabolic remodeling. Moreover, we demonstrate that hepatocytes are equipped with a flexible metabolic machinery able to adapt dynamically to changes during tissue regeneration.


Asunto(s)
Hepatocitos/metabolismo , Regeneración Hepática/fisiología , Hígado/metabolismo , Mitocondrias/metabolismo , Animales , Metabolómica/métodos , Ácido Pirúvico/metabolismo
13.
Cell Cycle ; 15(22): 3070-3081, 2016 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-27657745

RESUMEN

Cyclin A2 is an essential gene for development and in haematopoietic stem cells and therefore its functions in definitive erythropoiesis have not been investigated. We have ablated cyclin A2 in committed erythroid progenitors in vivo using erythropoietin receptor promoter-driven Cre, which revealed its critical role in regulating erythrocyte morphology and numbers. Erythroid-specific cyclin A2 knockout mice are viable but displayed increased mean erythrocyte volume and reduced erythrocyte counts, as well as increased frequency of erythrocytes containing Howell-Jolly bodies. Erythroblasts lacking cyclin A2 displayed defective enucleation, resulting in reduced production of enucleated erythrocytes and increased frequencies of erythrocytes containing nuclear remnants. Deletion of the Cdk inhibitor p27Kip1 but not Cdk2, ameliorated the erythroid defects resulting from deficiency of cyclin A2, confirming the critical role of cyclin A2/Cdk activity in erythroid development. Loss of cyclin A2 in bone marrow cells in semisolid culture prevented the formation of BFU-E but not CFU-E colonies, uncovering its essential role in BFU-E function. Our data unveils the critical functions of cyclin A2 in regulating mammalian erythropoiesis.


Asunto(s)
Forma de la Célula , Ciclina A2/metabolismo , Eritrocitos/citología , Eritrocitos/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Bromodesoxiuridina/metabolismo , Recuento de Células , Ciclo Celular , Núcleo Celular/metabolismo , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Células Eritroides/citología , Células Eritroides/metabolismo , Eritropoyesis , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/metabolismo , Ratones Endogámicos C57BL , Fenotipo , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo
14.
Cancers (Basel) ; 6(1): 79-111, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24419005

RESUMEN

The liver acts as a hub for metabolic reactions to keep a homeostatic balance during development and growth. The process of liver cancer development, although poorly understood, is related to different etiologic factors like toxins, alcohol, or viral infection. At the molecular level, liver cancer is characterized by a disruption of cell cycle regulation through many molecular mechanisms. In this review, we focus on the mechanisms underlying the lack of regulation of the cell cycle during liver cancer, focusing mainly on hepatocellular carcinoma (HCC). We also provide a brief summary of novel therapies connected to cell cycle regulation.

15.
Curr Biol ; 24(6): 598-608, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24613310

RESUMEN

BACKGROUND: Regulation of cell size requires coordination of growth and proliferation. Conditional loss of cyclin-dependent kinase 1 in mice permits hepatocyte growth without cell division, allowing us to study cell size in vivo using transcriptomics and metabolomics. RESULTS: Larger cells displayed increased expression of cytoskeletal genes but unexpectedly repressed expression of many genes involved in mitochondrial functions. This effect appears to be cell autonomous because cultured Drosophila cells induced to increase cell size displayed a similar gene-expression pattern. Larger hepatocytes also displayed a reduction in the expression of lipogenic transcription factors, especially sterol-regulatory element binding proteins. Inhibition of mitochondrial functions and lipid biosynthesis, which is dependent on mitochondrial metabolism, increased the cell size with reciprocal effects on cell proliferation in several cell lines. CONCLUSIONS: We uncover that large cell-size increase is accompanied by downregulation of mitochondrial gene expression, similar to that observed in diabetic individuals. Mitochondrial metabolism and lipid synthesis are used to couple cell size and cell proliferation. This regulatory mechanism may provide a possible mechanism for sensing metazoan cell size.


Asunto(s)
Tamaño de la Célula , Metaboloma/fisiología , Transcripción Genética/fisiología , Aerobiosis/fisiología , Animales , Proliferación Celular , Citoesqueleto/fisiología , Femenino , Regulación de la Expresión Génica/fisiología , Glucólisis/fisiología , Metabolismo de los Lípidos/fisiología , Hígado/citología , Ratones , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA