Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 325(6): G582-G592, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37847682

RESUMEN

Zinc fingers and homeoboxes 2 (Zhx2) are transcriptional regulators of liver gene expression with key functions in embryonic development as well as tissue regeneration in response to damage and disease, presumably through its control of target genes. Previous microarray data suggested that elongation of very long chain fatty acids-3 (Elovl3), a member of the ELOVL family of enzymes that synthesize very long chain fatty acids (VLCFAs), is a putative Zhx2 target gene. VLCFAs are core component of ceramides and other bioactive sphingolipids that are often dysregulated in diseases and regulate key cellular processes including proliferation. Since several previously identified Zhx2 targets become dysregulated in liver damage, we investigated the relationship between Zhx2 and Elovl3 in liver development, damage, and regeneration. Here, using mouse and cell models, we demonstrate that Zhx2 positively regulates Elovl3 expression in the liver and that male-biased hepatic Elovl3 expression is established between 4 and 8 wk of age in mice. Elovl3 is dramatically repressed in mouse models of liver regeneration, and the reduced Elovl3 levels in the regenerating liver are associated with changes in hepatic VLCFAs. Human hepatoma cell lines with forced Elovl3 expression have lower rates of cell growth; analysis of synchronized cells indicates that this reduced proliferation correlates with cells stalling in S-phase and lower mRNA levels of cell cyclins. Taken together, these data indicate that Elovl3 expression helps regulate cellular proliferation during liver development and regeneration, possibly through control of VLCFAs.NEW & NOTEWORTHY Numerous targets of the transcription factor Zhx2 are dysregulated in liver disease. We show that the elongase Elovl3 is a novel Zhx2 target. Elovl3 and Zhx2 expression change during liver regeneration, which is associated with changes in very long chain fatty acids. Forced Elovl3 expression reduces cell growth and blocks cell cycle progression. This suggests that Elovl3 may account, at least in part, for the relationship between Zhx2 and proliferation during liver development and disease.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Masculino , Ratones , Humanos , Animales , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Ácidos Grasos , Ciclo Celular , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/genética
2.
Hepatol Commun ; 6(12): 3550-3562, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36194180

RESUMEN

Liver cancer, comprised primarily of hepatocellular carcinoma (HCC), is the third leading cause of cancer deaths worldwide and increasing in Western countries. We previously identified the transcription factor zinc fingers and homeoboxes 2 (Zhx2) as a regulator of hepatic gene expression, and many Zhx2 target genes are dysregulated in HCC. Here, we investigate HCC in Zhx2-deficient mice using the diethylnitrosamine (DEN)-induced liver tumor model. Our study using whole-body Zhx2 knockout (Zhx2KO ) mice revealed the complete absence of liver tumors 9 and 10 months after DEN exposure. Analysis soon after DEN treatment showed no differences in expression of the DEN bioactivating enzyme cytochrome P450 2E1 (CYP2E1) and DNA polymerase delta 2, or in the numbers of phosphorylated histone variant H2AX foci between Zhx2KO and wild-type (Zhx2wt ) mice. The absence of Zhx2, therefore, did not alter DEN bioactivation or DNA damage. Zhx2KO livers showed fewer positive foci for Ki67 staining and reduced interleukin-6 and AKT serine/threonine kinase 2 expression compared with Zhx2wt livers, suggesting that Zhx2 loss reduces liver cell proliferation and may account for reduced tumor formation. Tumors were reduced but not absent in DEN-treated liver-specific Zhx2 knockout mice, suggesting that Zhx2 acts in both hepatocytes and nonparenchymal cells to inhibit tumor formation. Analysis of data from the Cancer Genome Atlas and Clinical Proteomic Tumor Consortium indicated that ZHX2 messenger RNA and protein levels were significantly higher in patients with HCC and associated with clinical pathological parameters. Conclusion: In contrast to previous studies in human hepatoma cell lines and other HCC mouse models showing that Zhx2 acts as a tumor suppressor, our data indicate that Zhx2 acts as an oncogene in the DEN-induced HCC model and is consistent with the higher ZHX2 expression in patients with HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Ratones , Carcinoma Hepatocelular/inducido químicamente , Dietilnitrosamina/efectos adversos , Genes Homeobox , Proteínas de Homeodominio/genética , Neoplasias Hepáticas/inducido químicamente , Ratones Endogámicos C57BL , Ratones Noqueados , Proteómica , Factores de Transcripción/genética , Dedos de Zinc
3.
Am J Physiol Endocrinol Metab ; 320(3): E609-E618, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33459178

RESUMEN

Obesity is associated with alterations in hepatic lipid metabolism. We previously identified the prorenin receptor (PRR) as a potential contributor to liver steatosis. Therefore, we aimed to determine the relative contribution of PRR and its soluble form, sPRR, to lipid homeostasis. PRR-floxed male mice were treated with an adeno-associated virus with thyroxine-binding globulin promoter-driven Cre to delete PRR in the liver [liver PRR knockout (KO) mice]. Hepatic PRR deletion did not change the body weight but increased liver weights. The deletion of PRR in the liver decreased peroxisome proliferator-activated receptor gamma (PPARγ) and triglyceride levels, but liver PRR KO mice exhibited higher plasma cholesterol levels and lower hepatic low-density lipoprotein receptor (LDLR) and Sortilin 1 (SORT1) proteins than control (CTL) mice. Surprisingly, hepatic PRR deletion elevated hepatic cholesterol, and up-regulated hepatic sterol regulatory element-binding protein 2 (SREBP2) and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG CoA-R) genes. In addition, the plasma levels of sPRR were significantly higher in liver PRR KO mice than in controls. In vitro studies in HepG2 cells demonstrated that sPRR treatment upregulated SREBP2, suggesting that sPRR could contribute to hepatic cholesterol biosynthesis. Interestingly, PRR, total cleaved and noncleaved sPRR contents, furin, and Site-1 protease (S1P) were elevated in the adipose tissue of liver PRR KO mice, suggesting that adipose tissue could contribute to the circulating pool of sPRR. Overall, this work supports previous works and opens a new area of investigation concerning the function of sPRR in lipid metabolism and adipose tissue-liver cross talk.NEW & NOTEWORTHY Hepatic PRR and its soluble form, sPRR, contribute to triglyceride and cholesterol homeostasis and hepatic inflammation. Deletion of hepatic PRR decreased triglyceride levels through a PRR-PPARγ-dependent mechanism but increased hepatic cholesterol synthesis through sPRR-medicated upregulation of SREBP-2. Our study highlighted a new paradigm of cross talk between the liver and the adipose tissue involving cholesterol and sPRR.


Asunto(s)
Homeostasis/genética , Metabolismo de los Lípidos/genética , Receptores de Superficie Celular/fisiología , Tejido Adiposo/metabolismo , Animales , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Células Hep G2 , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Solubilidad , Triglicéridos/metabolismo , Receptor de Prorenina
4.
Gene ; 767: 145162, 2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-32987105

RESUMEN

The mammalian Cytochrome P450 (Cyp) gene superfamily encodes enzymes involved in numerous metabolic pathways and are frequently expressed in the liver. Despite the remarkably high sequence similarity of Cyp2a4 and Cyp2a5 genes and their surrounding genomic regions, they exhibit differences in expression in the adult mouse liver. For example, Cyp2a4 is highly female-biased whereas Cyp2a5 is only moderately female-biased and Cyp2a4, but not Cyp2a5, is activated in liver cancer. We hypothesized that the limited sequence differences may help us identify the basis for this differential expression. An antisense expressed sequence tag had been uniquely annotated to the Cyp2a4 gene which led us to investigate this transcript as a possible regulator of this gene. We characterized the full-length antisense transcript and also discovered a similar transcript in the Cyp2a5 gene. These transcripts are nuclear long noncoding RNAs that are expressed similarly to their sense mRNA counterparts. This includes the sex-biased and liver tumor differences seen between the Cyp2a4 and Cyp2a5 genes, but we also find that these two genes and their antisense transcripts are expressed within different zones of the liver structure. Interestingly, while the differences in sex-biased expression of the mRNAs are established 1-2 months after birth, the antisense transcripts exhibit these expression differences earlier, at 3-4 weeks after birth. By analyzing published genomic data, we have identified candidate transcription factor binding sites that could account for differences in Cyp2a4/Cyp2a5 expression. Taken together, these studies characterize the first antisense RNAs within the Cyp supergene family and identify potential transcriptional and post-transcriptional mechanisms governing different Cyp2a4 and Cyp2a5 expression patterns in mouse liver.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/genética , Familia 2 del Citocromo P450/genética , Hígado/metabolismo , Esteroide Hidroxilasas/genética , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Familia 2 del Citocromo P450/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , Esteroide Hidroxilasas/metabolismo
5.
Genome Biol Evol ; 12(3): 174-184, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32125369

RESUMEN

The Zinc Fingers and Homeoboxes (Zhx) proteins, Zhx1, Zhx2, and Zhx3, comprise a small family of proteins containing two amino-terminal C2-H2 zinc fingers and four or five carboxy-terminal homeodomains. These multiple homeodomains make Zhx proteins unusual because the majority of homeodomain-containing proteins contain a single homeodomain. Studies in cultured cells and mice suggest that Zhx proteins can function as positive or negative transcriptional regulators. Zhx2 regulates numerous hepatic genes, and all three Zhx proteins have been implicated in different cancers. Because Zhx proteins contain multiple predicted homeodomains, are associated with interesting physiological traits, and seem to be only present in the vertebrate lineage, we investigated the evolutionary history of this small family by comparing Zhx homologs from a wide range of chordates. This analysis indicates that the zinc finger motifs and homeodomains are highly similar among all Zhx proteins and also identifies additional Zhx-specific conserved regions, including a 13 amino acid amino-terminal motif that is nearly identical among all gnathostome Zhx proteins. We found single Zhx proteins in the sea lamprey (Petromyzon marinus) and in the nonvertebrate chordates sea squirt (Ciona intestinalis) and lancelet (Branchiostoma floridae); these Zhx proteins are most similar to gnathostome Zhx3. Based on our analyses, we propose that a duplication of the primordial Zhx gene gave rise to Zhx3 and the precursor to Zhx1 and Zhx2. A subsequent tandem duplication of this precursor generated Zhx1 and Zhx2 found in gnathostomes.


Asunto(s)
Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Cordados/genética , Secuencia Conservada , Evolución Molecular , Proteínas de Homeodominio/clasificación , Humanos , Familia de Multigenes , Filogenia , Dominios Proteicos , Factores de Transcripción/clasificación
6.
J Med Chem ; 62(24): 11348-11358, 2019 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-31774672

RESUMEN

The importance of upregulated Wnt signaling in colorectal cancers led to efforts to develop inhibitors that target ß-catenin in this pathway. We now report that several "Wnt inhibitors" that allegedly target ß-catenin actually function as mitochondrial proton uncouplers that independently activate AMPK and concomitantly inhibit Wnt signaling. As expected for a process in which mitochondrial uncoupling diminishes ATP production, a mitochondrial proton uncoupler, FCCP, and a glucose metabolic inhibitor, 2-DG, activated AMPK and inhibited Wnt signaling. Also consistent with these findings, a well-known "Wnt inhibitor", FH535, functioned as a proton uncoupler, and in support of this finding, the N-methylated analog, 2,5-dichloro-N-methyl-N-(2-methyl-4-nitrophenyl)benzenesulfonamide (FH535-M), was inactive as an uncoupler and Wnt inhibitor. Apart from suggesting an opportunity to develop dual Wnt inhibitors and AMPK activators, these findings provide a cautionary tale that claims for Wnt inhibition alone require scrutiny as possible mitochondrial proton uncouplers or inhibitors of the electron transport chain.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Encéfalo/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Activadores de Enzimas/farmacología , Hidrocarburos Fluorados/farmacología , Mitocondrias/efectos de los fármacos , Urea/farmacología , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Encéfalo/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Metabolismo Energético , Activación Enzimática , Activadores de Enzimas/química , Regulación Neoplásica de la Expresión Génica , Humanos , Hidrocarburos Fluorados/química , Mitocondrias/metabolismo , Consumo de Oxígeno , Sulfonamidas/química , Sulfonamidas/farmacología , Células Tumorales Cultivadas , Urea/análogos & derivados , Urea/química
7.
Mamm Genome ; 30(7-8): 226-236, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31321500

RESUMEN

BALB/cJ mice exhibit considerable phenotypic differences with other BALB/c substrains. Some of these traits involve the liver, including persistent postnatal expression of genes that are normally expressed only in the fetal liver and reduced expression of major urinary proteins. These traits are due to a mutation that dramatically reduces expression of the gene encoding the transcription factor Zinc fingers and homeoboxes 2 (Zhx2). BALB/cJ mice also exhibit reduced serum lipid levels and resistance to atherosclerosis compared to other mouse strains when placed on a high-fat diet. This trait is also due, at least in part, to the Zhx2 mutation. Microarray analysis identified many genes affecting lipid homeostasis, including Lipoprotein lipase, that are dysregulated in BALB/cJ liver. This led us to investigate whether hepatic lipid levels would be different between BALB/cJ and BALB/c mice when placed on a normal chow or a high-fat chow diet. On the high-fat chow, BALB/cJ mice had increased weight gain, increased liver:body weight ratio, elevated hepatic lipid accumulation and markers of liver damage when compared to BALB/c mice. These traits in BALB/cJ mice were only partially reversed by a hepatocyte-specific Zhx2 transgene. These data indicate that Zhx2 reduces liver lipid levels and is hepatoprotective in mice on a high-fat diet, but the partial rescue by the Zhx2 transgene suggests a contribution by both parenchymal and non-parenchymal cells. A model to account for the cardiovascular and liver phenotype in mice with reduced Zhx2 levels is provided.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Proteínas de Homeodominio/genética , Metabolismo de los Lípidos/genética , Hígado/patología , Alanina Transaminasa/sangre , Animales , Femenino , Hepatocitos/metabolismo , Hepatocitos/patología , Lípidos/sangre , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mutación , Aumento de Peso/genética
8.
Int J Cancer ; 143(12): 3120-3130, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752719

RESUMEN

Initiation of hepatocellular carcinoma (HCC) by chronic hepatitis B virus (HBV) infection is a complex process that includes both oncogene activation and tumor suppressor inhibition. The HBV X (HBx) protein has an important and complex role in processes leading to HCC. We previously identified the mammalian Zinc fingers and homeoboxes 2 (ZHX2) gene as an HCC-associated tumor suppressor gene. In the present study, we investigated whether the oncogenic properties of HBV and, more specifically, HBx, involved ZHX2 silencing. Our data indicates that ZHX2 expression is significantly decreased in tumor tissues from HBV-positive HCC patients and livers from HBV transgenic mice. In vitro and in vivo studies confirmed that HBV-encoded proteins, particularly HBx, inhibits both the expression and tumor suppression properties of ZHX2. Further analyses identified miR-155, a well-known oncomiR in various cancers, as an important link between HBx and ZHX2 inhibition. Increased miR-155 levels were found in HBV-positive tumors, livers of HBV transgenic mice and HBx-overexpressing hepatoma cell lines. MiR-155 overexpression reduced ZHX2 levels via miR-155 seed sites in the ZHX2 3'UTR, whereas blocking miR-155 levels led to increased ZHX2 levels. Taken together, our data indicate that HCC-promoting properties of HBV may include ZHX2 silencing via a miR-155 dependent pathway and suggests a novel therapy for HBV-related HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Proliferación Celular/genética , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/complicaciones , Proteínas de Homeodominio/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , MicroARNs/genética , Factores de Transcripción/metabolismo , Adulto , Anciano , Animales , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Silenciador del Gen , Proteínas de Homeodominio/genética , Humanos , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Persona de Mediana Edad , Factores de Riesgo , Transactivadores/metabolismo , Factores de Transcripción/genética , Regulación hacia Arriba , Proteínas Reguladoras y Accesorias Virales
9.
J Biol Chem ; 292(16): 6765-6774, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28258223

RESUMEN

The mouse major urinary proteins (Mups) are encoded by a large family of highly related genes clustered on chromosome 4. Mups, synthesized primarily and abundantly in the liver and secreted through the kidneys, exhibit male-biased expression. Mups bind a variety of volatile ligands; these ligands, and Mup proteins themselves, influence numerous behavioral traits. Although urinary Mup protein levels vary between inbred mouse strains, this difference is most pronounced in BALB/cJ mice, which have dramatically low urinary Mup levels; this BALB/cJ trait had been mapped to a locus on chromosome 15. We previously identified Zhx2 (zinc fingers and homeoboxes 2) as a regulator of numerous liver-enriched genes. Zhx2 is located on chromosome 15, and a natural hypomorphic mutation in the BALB/cJ Zhx2 allele dramatically reduces Zhx2 expression. Based on these data, we hypothesized that reduced Zhx2 levels are responsible for lower Mup expression in BALB/cJ mice. Using both transgenic and knock-out mice along with in vitro assays, our data show that Zhx2 binds Mup promoters and is required for high levels of Mup expression in the adult liver. In contrast to previously identified Zhx2 targets that appear to be repressed by Zhx2, Mup genes are positively regulated by Zhx2. These data identify Zhx2 as a novel regulator of Mup expression and indicate that Zhx2 activates as well as represses expression of target genes.


Asunto(s)
Proteínas de Homeodominio/fisiología , Hígado/metabolismo , Proteínas/fisiología , Factores de Transcripción/fisiología , Alelos , Animales , Línea Celular , Cromatina/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Riñón/metabolismo , Ligandos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica
10.
Gene Expr ; 17(1): 7-17, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27197076

RESUMEN

The mammalian cytochrome P450 (Cyp) gene family encodes a large number of structurally related enzymes that catalyze a variety of metabolic and detoxification reactions. The liver is the primary site of Cyp expression in terms of expression levels and number of expressed genes, consistent with this organ's essential role in metabolism of endogenous and xenobiotic compounds. Many Cyp genes exhibit sexually dimorphic expression. For example, Cyp2a4 is expressed significantly higher in the adult liver of female mice compared to male mice. An exception to this pattern is seen in BALB/cJ mice, where male hepatic Cyp2a4 mRNA levels are substantially elevated compared to male mice of other strains. The Zinc fingers and homeoboxes 2 (Zhx2) protein governs the silencing of several genes in the postnatal liver, including α-fetoprotein, H19, and glypican 3. Zhx2 also regulates numerous hepatic genes that govern lipid homeostasis. We previously showed that the Zhx2 gene is mutated in BALB/cJ mice, which led us to consider whether elevated male hepatic Cyp2a4 levels in this strain are due to this Zhx2 mutation. Using mice with a conditional Zhx2 deletion, we show here that the absence of Zhx2 in hepatocytes results in increased Cyp2a4 expression in adult male liver. We extend this finding to show that additional Cyp genes are disregulated in the absence of Zhx2. We also show that mRNA levels of Cyp2a4 and several other female-biased Cyp genes are increased, and male-biased Cyp4a12 is decreased in mouse liver tumors. These data indicate that Zhx2 is a novel regulator of sex-biased Cyp gene expression in the normal and diseased liver.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Hígado/metabolismo , Animales , Femenino , Glipicanos/metabolismo , Hepatocitos/metabolismo , Homeostasis/fisiología , Lípidos/fisiología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , ARN Mensajero/genética , Caracteres Sexuales , alfa-Fetoproteínas/metabolismo
11.
Sci Rep ; 5: 11979, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26173901

RESUMEN

Alpha-fetoprotein (AFP) represents a classical model system to study developmental gene regulation in mammalian cells. We previously reported that liver ZBTB20 is developmentally regulated and plays a central role in AFP postnatal repression. Here we show that ZBTB20 is a sequence-specific transcriptional repressor of AFP. By ELISA-based DNA-protein binding assay and conventional gel shift assay, we successfully identified a ZBTB20-binding site at -104/-86 of mouse AFP gene, flanked by two HNF1 sites and two C/EBP sites in the proximal promoter. Importantly, mutation of the core sequence in this site fully abolished its binding to ZBTB20 in vitro, as well as the repression of AFP promoter activity by ZBTB20. The unique ZBTB20 site was highly conserved in rat and human AFP genes, but absent in albumin genes. These help to explain the autonomous regulation of albumin and AFP genes in the liver after birth. Furthermore, we demonstrated that transcriptional repression of AFP gene by ZBTB20 was liver-specific. ZBTB20 was dispensable for AFP silencing in other tissues outside liver. Our data define a cognate ZBTB20 site in AFP promoter which mediates the postnatal repression of AFP gene in the liver.


Asunto(s)
Factores de Transcripción/metabolismo , alfa-Fetoproteínas/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , ADN/química , ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Genes Reporteros , Células Hep G2 , Humanos , Hígado/metabolismo , Ratones , Ratones Noqueados , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/química , Factores de Transcripción/genética , Transcripción Genética , alfa-Fetoproteínas/química , alfa-Fetoproteínas/genética
12.
Oncotarget ; 6(2): 1049-63, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25473899

RESUMEN

We previously reported the tumor suppressor function of Zinc-fingers and homeoboxes 2 (ZHX2) in hepatocellular carcinoma (HCC). Other studies indicate the association of increased ZHX2 expression with improved response to high dose chemotherapy in multiple myeloma. Here, we aim to test whether increased ZHX2 levels in HCC cells repress multidrug resistance 1(MDR1) expression resulting in increased sensitivity to chemotherapeutic drugs. We showed evidence that increased ZHX2 levels correlated with reduced MDR1 expression and enhanced the cytotoxicity of CDDP and ADM in different HCC cell lines. Consistently, elevated ZHX2 significantly reduced ADM efflux in HepG2 cells and greatly increased the CDDP-mediated suppression of liver tumor growth in vivo. Furthermore, immunohistochemical staining demonstrated the inverse correlation of ZHX2 and MDR1 expression in HCC tissues. Luciferase report assay showed that ZHX2 repressed the MDR1 promoter activity, while knockdown of NF-YA or mutating the NF-Y binding site eliminated this ZHX2-mediated repression of MDR1 transcription. Co-IP and ChIP assay further suggested that ZHX2 interacted with NF-YA and reduced NF-Y binding to the MDR1 promoter. Taken together, we clarify that ZHX2 represses NF-Y-mediated activation of MDR1 transcription and, in doing so, enhances the effects of chemotherapeutics in HCC cells both in vitro and in vivo.


Asunto(s)
Factor de Unión a CCAAT/genética , Carcinoma Hepatocelular/genética , Proteínas de Homeodominio/genética , Neoplasias Hepáticas/genética , Factores de Transcripción/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adulto , Anciano , Animales , Antineoplásicos/farmacología , Factor de Unión a CCAAT/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/farmacología , Femenino , Células HEK293 , Células Hep G2 , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
PLoS One ; 9(6): e99272, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24940873

RESUMEN

Activation of the Wnt/ß-catenin pathway has been observed in at least 1/3 of hepatocellular carcinomas (HCC), and a significant number of these have mutations in the ß-catenin gene. Therefore, effective inhibition of this pathway could provide a novel method to treat HCC. The purposed of this study was to determine whether FH535, which was previously shown to block the ß-catenin pathway, could inhibit ß-catenin activation of target genes and inhibit proliferation of Liver Cancer Stem Cells (LCSC) and HCC cell lines. Using ß-catenin responsive reporter genes, our data indicates that FH535 can inhibit target gene activation by endogenous and exogenously expressed ß-catenin, including the constitutively active form of ß-catenin that contains a Serine37Alanine mutation. Our data also indicate that proliferation of LCSC and HCC lines is inhibited by FH535 in a dose-dependent manner, and that this correlates with a decrease in the percentage of cells in S phase. Finally, we also show that expression of two well-characterized targets of ß-catenin, Cyclin D1 and Survivin, is reduced by FH535. Taken together, this data indicates that FH535 has potential therapeutic value in treatment of liver cancer. Importantly, these results suggest that this therapy may be effective at several levels by targeting both HCC and LCSC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/efectos de los fármacos , Sulfonamidas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Terapia Molecular Dirigida , Survivin , Activación Transcripcional/efectos de los fármacos
14.
Anticancer Res ; 34(4): 1709-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24692700

RESUMEN

BACKGROUND/AIM: The aim of this study is to find synergistic effect using FH535 and sorafenib by targeting the RAS/RAF/MAPK and WNT/ß-catenin pathways. MATERIALS AND METHODS: 3H-Thymidine incorporation assays were performed to address Huh7 and liver cancer stem cell (LCSC) inhibition using FH535 and sorafenib, alone and in combination. Calcusyn analysis was used to calculate the combination index (CI). A western blot assay was performed to check for potential targets. RESULTS: FH535 and sorafenib caused inhibition of Huh7 and LCSC. Combination therapy was significantly better than monotherapy in inhibition of HuH7. Combination with sorafenib and FH535 was found to be synergistic in inhibition of LCSC with a CI of less than 1. The western blot assay demonstrated enhanced cleaved poly (ADP-ribose) polymerase (PARP) and inhibition of cyclin D1, B-cell lymphoma 2 (Bcl2), survivin and cellular myelocytomatosis oncogene (c-MYC). CONCLUSION: FH535 and sorafenib combination produced synergistic effect on inhibition of HCC and LCSC. Our study demonstrated that FH535 can induce apoptosis in these two different hepatocellular carcinoma (HCC) cell lines.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Quinasas raf/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Inmunofenotipificación , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacología , Fenotipo , Compuestos de Fenilurea/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sorafenib , Sulfonamidas/farmacología , Survivin
15.
J Surg Res ; 185(1): 225-30, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23769634

RESUMEN

BACKGROUND: Deregulated Ras/Raf/mitogen-activated protein kinase and PI3 K/AKT/mTOR signaling pathways are significant in hepatocellular carcinoma proliferation (HCC). In this study we evaluated differences in the antiproliferative effect of dual PI3 K/Akt/mTOR and Ras/Raf/mitogen-activated protein kinase inhibition of non liver cancer stem cell lines (PLC and HuH7) and liver cancer stem cell (LCSC) lines (CD133, CD44, CD24, and aldehyde dehydrogenase 1-positive cells). MATERIALS AND METHODS: Flow cytometry was performed on the resulting tumors to identify the LCSC markers CD133, CD44, CD24, and aldehyde dehydrogenase 1. Methylthiazol tetrazolium assay was used to assess cellular proliferation. Finally, a Western blot assay was used to evaluate for inhibition of specific enzymes in these two signaling pathways. RESULTS: Using flow cytometry, we found that LCSC contain 64.4% CD133 + cells, 83.2% CD44 + cells, and 96.4% CD24 + cells. PKI-587 and sorafenib caused inhibiton of LCSC and HCC cell proliferation. PLC cells were more sensitive to PKI-587 than LCSC or Huh7 (P < 0.001). Interestingly, HuH7 cells were more sensitive to sorafenib than LCSC or PLC cells. Additionally, combination therapy with PKI-587 and sorafenib caused significantly more inhibition than monotherapy in HuH7, PLC, and LCSC. Using the methylthiazol tetrazolium assay, we found that the LCSC proliferation was inhibited with sorafenib monotherapy 39% at 5 µM (P < 0.001; n = 12) and 67% by PKI-587 at 0.1 µM (P = 0.002, n = 12) compared with control. The combination of PKI-587 and sorafenib, however, synergistically inhibited LCSC proliferation by 86% (P = 0.002; n = 12). CONCLUSIONS: LCSC (CD133+, CD44+, CD24+) were able to develop very aggressive tumors with low cell concentrations at 4 to 6 wk. Cells CD133+, CD44+, CD24+, which demonstrated at least moderate resistance to therapy in vitro. The combination of PKI-587 and sorafenib was better than either drug alone at inhibiting of LCSC and on HCC cell proliferation.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Morfolinas/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Triazinas/farmacología , Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Neoplásicas/citología , Niacinamida/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal/efectos de los fármacos , Sorafenib , Serina-Treonina Quinasas TOR/metabolismo , Quinasas raf/metabolismo
16.
J Am Assoc Lab Anim Sci ; 52(1): 17-21, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23562028

RESUMEN

The use of a murine nonsurgical embryo transfer (NSET) device had been described previously for the transfer of blastocysts, morulae, DNA-microinjected embryos, and embryonic stem cell-containing embryos to create genetically modified mice. However, physiologic effects of the NSET device and traditional surgical methods had not been compared directly. Here we used electrocardiography and fecal corticosterone levels to monitor pseudopregnant mice that underwent anesthesia only, the NSET procedure with or without anesthesia, or surgery. These procedures were performed without the use of actual embryos, to focus on effects of the procedures themselves rather than on any physiologic effects due to the deposition of embryos. As compared with surgery and anesthesia, the NSET procedure was associated with less fluctuation in cardiac rhythm and lower levels of the stress biomarker fecal corticosterone. These results indicate that use of the NSET device avoids these physi- ological perturbations as well as other disadvantages of surgery (for example, postoperative pain and need for postoperative analgesia) and therefore provides a valuable refinement of existing mouse embryo transfer procedures.


Asunto(s)
Transferencia de Embrión/veterinaria , Ratones Transgénicos/embriología , Anestesia General/veterinaria , Bienestar del Animal , Animales , Blastocisto , Transferencia de Embrión/métodos , Femenino , Ratones , Mórula , Embarazo
17.
Anticancer Res ; 32(7): 2531-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22753710

RESUMEN

BACKGROUND: Deregulated RAS/RAF/MAPK and PI3K/AKT/mTOR signaling pathways are found in hepatocellular carcinoma (HCC). This study aimed to test the inhibitory effects of PI-103 (a small molecule inhibitor of PI3K and mTOR) and sorafenib as single agents and in combination on HCC tumorigenesis in an in vivo xenograft model. MATERIALS AND METHODS: In vitro study: Huh7 proliferation was assayed by 3H-thymidine incorporation and by thiazolyl blue tetrazolium bromide (MTT) assay. Western blots were used to detect phosphorylation of the key enzymes in the two pathways. In vivo study: Human HCC cell line Huh7 was inoculated into nude mice s.c. and the mice were treated with sorafenib (20 mg/kg/day) and PI-103 (5 mg/kg, every 4 days). Tumor size was measured every other day. Tumors were isolated for western blot and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay detection of apoptosis and signaling pathway enzymes. RESULTS: Our in vitro study found that combination of sorafenib and PI-103 additively inhibited Huh7 proliferation as compared to single-agent treatment. Sorafenib and PI-103 as single agents differentially inhibited or activated key enzymes (MEK, ERK, AKT, mTOR, and S6K) in PI3K/AKT/mTOR and RAS/RAF/MAPK signaling pathways. Combination of sorafenib and PI-103 inhibited all the key enzymes in the two pathways. Our in vivo study demonstrated significant differences between control group, mono-drug groups and drug-combination group (p<0.05). Combination of Sorafenib and PI-103 more efficiently inhibited tumorigenesis as compared to mono-drug treatments (p<0.032). CONCLUSION: The combination of PI-103 and sorafenib has the advantage over mono-drug therapy on inhibition of HCC cell proliferation and tumorigenesis by inhibiting both PI3K/AKT/mTOR and RAS/RAF/MAPK signaling pathways.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Bencenosulfonatos/administración & dosificación , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Furanos/administración & dosificación , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Niacinamida/análogos & derivados , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/metabolismo , Compuestos de Fenilurea , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piridinas/administración & dosificación , Pirimidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Sorafenib , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Hepatology ; 56(5): 1892-901, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22544812

RESUMEN

UNLABELLED: We previously showed that mouse alpha-fetoprotein (AFP) enhancer 3 activity is highly restricted to pericentral hepatocytes in the adult liver. Here, using transgenic mice, we show that the upstream enhancer of the rat glutamine synthetase gene is also active, specifically in pericentral regions. Activity of both enhancers is lost in the absence of ß-catenin, a key regulator of zonal gene expression in the adult liver. Both enhancers contain a single, highly conserved T-cell factor/lymphoid enhancer factor binding site that is required for responsiveness to ß-catenin. We also show that endogenous AFP messenger RNA levels in the perinatal liver are lower when ß-catenin is reduced. CONCLUSION: These data identify the first distinct zonally active regulatory regions required for ß-catenin responsiveness in the adult liver, and suggest that postnatal AFP repression and the establishment of zonal regulation are controlled, at least in part, by the same factors.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Glutamato-Amoníaco Ligasa/genética , Hígado/enzimología , Transducción de Señal , alfa-Fetoproteínas/genética , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Regulación del Desarrollo de la Expresión Génica , Hígado/metabolismo , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo , Ratas , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Transfección , alfa-Fetoproteínas/metabolismo , beta Catenina/genética
19.
Gastroenterology ; 142(7): 1559-70.e2, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22406477

RESUMEN

BACKGROUND & AIMS: Zinc-fingers and homeoboxes 2 (ZHX2) represses transcription of several genes associated with liver cancer. However, little is known about the role of ZHX2 in the development of hepatocellular carcinoma (HCC). We investigated the mechanisms by which ZHX2 might affect proliferation of HCC cells. METHODS: We overexpressed and knocked down ZHX2 in HCC cells and analyzed the effects on proliferation, colony formation, and the cell cycle. We also analyzed the effects of ZHX2 overexpression in growth of HepG2.2.15 tumor xenografts in nude mice. Chromatin immunoprecipitation and luciferase reporter assays were used to measure binding of ZHX2 target promoters. Levels of ZHX2 in HCC samples were evaluated by immunohistochemistry. RESULTS: ZHX2 overexpression significantly reduced proliferation of HCC cells and growth of tumor xenografts in mice; it led to G1 arrest and reduced levels of Cyclins A and E in HCC cell lines. ZHX2 bound to promoter regions of CCNA2 (which encodes Cyclin A) and CCNE1 (which encodes Cyclin E) and inhibited their transcription. Knockdown of Cyclin A or Cyclin E reduced the increased proliferation mediated by ZHX2 knockdown. Nuclear localization of ZHX2 was required for it to inhibit proliferation of HCC cells in culture and in mice. Nuclear localization of ZHX2 was reduced in human HCC samples, even in small tumors (diameter, <5 cm), compared with adjacent nontumor tissues. Moreover, reduced nuclear levels of ZHX2 correlated with reduced survival times of patients, high levels of tumor microvascularization, and hepatocyte proliferation. CONCLUSIONS: ZHX2 inhibits HCC cell proliferation by preventing expression of Cyclins A and E, and reduces growth of xenograft tumors in mice. Loss of nuclear ZHX2 might be an early step in the development of HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Proliferación Celular , Proteínas de Homeodominio/metabolismo , Neoplasias Hepáticas/patología , Animales , Carcinoma Hepatocelular/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina A/metabolismo , Ciclina E/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/farmacología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Activación Transcripcional/efectos de los fármacos
20.
J Environ Pathol Toxicol Oncol ; 30(4): 273-82, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22181977

RESUMEN

We report the role of dietary glycine and the type of oil used as a vehicle in the hepatotoxicity of control rats and rats treated with 2,2',4,4',5,5'-hexachlorobiphenyl (PCB-153). In our first experiment, glycine or valine (as control) was fed in an unrefined diet at 5% for the entire study duration (5 days) to inhibit Kupffer cell activity. PCB-153 (100 or 300 µmol/kg) dissolved in medium chain triglyceride (MCT) oil, was injected intraperitoneally 2 days before euthanasia; the peroxisome proliferator Wy-14,643 was included as a positive control. MCT oil decreased cell proliferation by approximately 50%. PCB-153 slightly increased hepatic cell proliferation, but dietary glycine did not reduce cell proliferation. Because of the inhibition of cell proliferation in rats receiving MCT oil compared with rats receiving no injection, we hypothesized that MCT oil may have been inhibiting the hepatocyte proliferation in PCB-153-treated rats. We therefore performed another experiment using 3 types of oil as a vehicle for PCB-153: MCT oil, corn oil, and olive oil. Rats were injected with PCB-153 (300 µmol/kg) or one of the vehicles, again 2 days before euthanasia. MCT oil again decreased the hepatocyte proliferation by approximately 50%. In rats receiving PCB-153, hepatocyte proliferation was slightly higher than their respective vehicle controls for corn oil and olive oil but not for MCT oil. These studies show that the oil vehicle is important in cell proliferation after PCB exposure, with MCT oil appearing to be protective.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glicina/farmacología , Hígado/efectos de los fármacos , Aceites/farmacología , Vehículos Farmacéuticos/farmacología , Bifenilos Policlorados/toxicidad , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Aceite de Maíz/farmacología , Citocromo P-450 CYP2B1/metabolismo , Hígado/patología , Masculino , Aceite de Oliva , Aceites de Plantas/farmacología , Bifenilos Policlorados/química , Ratas , Ratas Sprague-Dawley , Esteroide Hidroxilasas/metabolismo , Triglicéridos/química , Triglicéridos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...