Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
bioRxiv ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38464291

RESUMEN

Lung cancer, the leading cause of cancer mortality, exhibits diverse histological subtypes and genetic complexities. Numerous preclinical mouse models have been developed to study lung cancer, but data from these models are disparate, siloed, and difficult to compare in a centralized fashion. Here we established the Lung Cancer Mouse Model Database (LCMMDB), an extensive repository of 1,354 samples from 77 transcriptomic datasets covering 974 samples from genetically engineered mouse models (GEMMs), 368 samples from carcinogen-induced models, and 12 samples from a spontaneous model. Meticulous curation and collaboration with data depositors have produced a robust and comprehensive database, enhancing the fidelity of the genetic landscape it depicts. The LCMMDB aligns 859 tumors from GEMMs with human lung cancer mutations, enabling comparative analysis and revealing a pressing need to broaden the diversity of genetic aberrations modeled in GEMMs. Accompanying this resource, we developed a web application that offers researchers intuitive tools for in-depth gene expression analysis. With standardized reprocessing of gene expression data, the LCMMDB serves as a powerful platform for cross-study comparison and lays the groundwork for future research, aiming to bridge the gap between mouse models and human lung cancer for improved translational relevance.

2.
Antioxidants (Basel) ; 10(2)2021 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-33557356

RESUMEN

Dietary antioxidants and supplements are widely used to protect against cancer, even though it is now clear that antioxidants can promote tumor progression by helping cancer cells to overcome barriers of oxidative stress. Although recent studies have, in great detail, explored the role of antioxidants in lung and skin tumors driven by RAS and RAF mutations, little is known about the impact of antioxidant supplementation on other cancers, including Wnt-driven tumors originating from the gut. Here, we show that supplementation with the antioxidants N-acetylcysteine (NAC) and vitamin E promotes intestinal tumor progression in the ApcMin mouse model for familial adenomatous polyposis, a hereditary form of colorectal cancer, driven by Wnt signaling. Both antioxidants increased tumor size in early neoplasias and tumor grades in more advanced lesions without any impact on tumor initiation. Importantly, NAC treatment accelerated tumor progression at plasma concentrations comparable to those obtained in human subjects after prescription doses of the drug. These results demonstrate that antioxidants play an important role in the progression of intestinal tumors, which may have implications for patients with or predisposed to colorectal cancer.

4.
Sci Rep ; 10(1): 14156, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843651

RESUMEN

Recent data suggest that the transcription factor Zfp148 represses activation of the tumor suppressor p53 in mice and that therapeutic targeting of the human orthologue ZNF148 could activate the p53 pathway without causing detrimental side effects. We have previously shown that Zfp148 deficiency promotes p53-dependent proliferation arrest of mouse embryonic fibroblasts (MEFs), but the underlying mechanism is not clear. Here, we showed that Zfp148 deficiency downregulated cell cycle genes in MEFs in a p53-dependent manner. Proliferation arrest of Zfp148-deficient cells required increased expression of ARF, a potent activator of the p53 pathway. Chromatin immunoprecipitation showed that Zfp148 bound to the ARF promoter, suggesting that Zfp148 represses ARF transcription. However, Zfp148 preferentially bound to promoters of other transcription factors, indicating that deletion of Zfp148 may have pleiotropic effects that activate ARF and p53 indirectly. In line with this, we found no evidence of genetic interaction between TP53 and ZNF148 in CRISPR and siRNA screen data from hundreds of human cancer cell lines. We conclude that Zfp148 deficiency, by increasing ARF transcription, downregulates cell cycle genes and cell proliferation in a p53-dependent manner. However, the lack of genetic interaction between ZNF148 and TP53 in human cancer cells suggests that therapeutic targeting of ZNF148 may not increase p53 activity in humans.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Transducción de Señal/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/fisiología , Animales , Sistemas CRISPR-Cas , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular , Línea Celular , Inmunoprecipitación de Cromatina , Cisplatino/toxicidad , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/fisiología , Regulación hacia Abajo , Factores de Transcripción E2F/fisiología , Etopósido/toxicidad , Fibroblastos , Ontología de Genes , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología
5.
Sci Rep ; 9(1): 14667, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604991

RESUMEN

The mut-T homolog-1 (MTH1) inhibitor TH588 has shown promise in preclinical cancer studies but its targeting specificity has been questioned. Alternative mechanisms for the anti-cancer effects of TH588 have been suggested but the question remains unresolved. Here, we performed an unbiased CRISPR screen on human lung cancer cells to identify potential mechanisms behind the cytotoxic effect of TH588. The screen identified pathways and complexes involved in mitotic spindle regulation. Using immunofluorescence and live cell imaging, we showed that TH588 rapidly reduced microtubule plus-end mobility, disrupted mitotic spindles, and prolonged mitosis in a concentration-dependent but MTH1-independent manner. These effects activated a USP28-p53 pathway - the mitotic surveillance pathway - that blocked cell cycle reentry after prolonged mitosis; USP28 acted upstream of p53 to arrest TH588-treated cells in the G1-phase of the cell cycle. We conclude that TH588 is a microtubule-modulating agent that activates the mitotic surveillance pathway and thus prevents cancer cells from re-entering the cell cycle.


Asunto(s)
Carcinoma de Células Grandes/tratamiento farmacológico , Enzimas Reparadoras del ADN/genética , Monoéster Fosfórico Hidrolasas/genética , Pirimidinas/farmacología , Ubiquitina Tiolesterasa/genética , Antineoplásicos/farmacología , Sistemas CRISPR-Cas/genética , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Microtúbulos/efectos de los fármacos , Mitosis/efectos de los fármacos , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Huso Acromático/efectos de los fármacos , Moduladores de Tubulina/farmacología , Proteína p53 Supresora de Tumor/genética
6.
Sci Rep ; 9(1): 15385, 2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31659186

RESUMEN

The search for biomarkers associated with obesity-related diseases is ongoing, but it is not clear whether plasma and serum can be used interchangeably in this process. Here we used high-throughput screening to analyze 358 proteins and 76 lipids, selected because of their relevance to obesity-associated diseases, in plasma and serum from age- and sex-matched lean and obese humans. Most of the proteins/lipids had similar concentrations in plasma and serum, but a subset showed significant differences. Notably, a key marker of cardiovascular disease PAI-1 showed a difference in concentration between the obese and lean groups only in plasma. Furthermore, some biomarkers showed poor correlations between plasma and serum, including PCSK9, an important regulator of cholesterol homeostasis. Collectively, our results show that the choice of biofluid may impact study outcome when screening for obesity-related biomarkers and we identify several markers where this will be the case.


Asunto(s)
Enfermedades Renales/sangre , Síndrome Metabólico/sangre , Obesidad/sangre , Inhibidor 1 de Activador Plasminogénico/sangre , Proproteína Convertasa 9/sangre , Adulto , Biomarcadores/sangre , Femenino , Humanos , Enfermedades Renales/complicaciones , Masculino , Síndrome Metabólico/complicaciones , Persona de Mediana Edad , Obesidad/complicaciones , Inhibidor 1 de Activador Plasminogénico/normas , Proproteína Convertasa 9/normas
7.
Cell ; 178(2): 330-345.e22, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31257027

RESUMEN

For tumors to progress efficiently, cancer cells must overcome barriers of oxidative stress. Although dietary antioxidant supplementation or activation of endogenous antioxidants by NRF2 reduces oxidative stress and promotes early lung tumor progression, little is known about its effect on lung cancer metastasis. Here, we show that long-term supplementation with the antioxidants N-acetylcysteine and vitamin E promotes KRAS-driven lung cancer metastasis. The antioxidants stimulate metastasis by reducing levels of free heme and stabilizing the transcription factor BACH1. BACH1 activates transcription of Hexokinase 2 and Gapdh and increases glucose uptake, glycolysis rates, and lactate secretion, thereby stimulating glycolysis-dependent metastasis of mouse and human lung cancer cells. Targeting BACH1 normalized glycolysis and prevented antioxidant-induced metastasis, while increasing endogenous BACH1 expression stimulated glycolysis and promoted metastasis, also in the absence of antioxidants. We conclude that BACH1 stimulates glycolysis-dependent lung cancer metastasis and that BACH1 is activated under conditions of reduced oxidative stress.


Asunto(s)
Antioxidantes/farmacología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Glucólisis/efectos de los fármacos , Neoplasias Pulmonares/patología , Animales , Antioxidantes/administración & dosificación , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Movimiento Celular/efectos de los fármacos , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Hemo/metabolismo , Hexoquinasa/antagonistas & inhibidores , Hexoquinasa/genética , Hexoquinasa/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Factor 2 Relacionado con NF-E2/metabolismo , Metástasis de la Neoplasia , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
PLoS One ; 11(12): e0166918, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27973542

RESUMEN

BACKGROUND: Vascularization of atherosclerotic plaques has been linked to plaque vulnerability. The aim of this study was to test if the vascularization was increased in upstream regions of early atherosclerotic carotid plaques and also to test if the same pattern of vascularization was seen in complicated, symptomatic plaques. METHODS: We enrolled 45 subjects with early atherosclerotic lesions for contrast enhanced ultrasound and evaluated the percentage of plaque area in a longitudinal ultrasound section which contained contrast agent. Contrast-agent uptake was evaluated in both the upstream and downstream regions of the plaque. We also collected carotid endarterectomy specimens from 56 subjects and upstream and downstream regions were localized using magnetic resonance angiography and analyzed using histopathology and immunohistochemistry. RESULTS: Vascularization was increased in the upstream regions of early carotid plaques compared with downstream regions (30% vs. 23%, p = 0.033). Vascularization was also increased in the upstream regions of advanced atherosclerotic lesions compared with downstream regions (4.6 vs. 1.4 vessels/mm2, p = 0.001) and was associated with intra-plaque hemorrhage and inflammation. CONCLUSIONS: Vascularization is increased in the upstream regions of both early and advanced plaques and is in advanced lesions mainly driven by inflammation.


Asunto(s)
Enfermedades de las Arterias Carótidas/patología , Neovascularización Patológica/patología , Placa Aterosclerótica/patología , Anciano , Arterias Carótidas/patología , Estenosis Carotídea/patología , Estudios Transversales , Progresión de la Enfermedad , Endarterectomía Carotidea , Femenino , Humanos , Inmunohistoquímica , Inflamación/patología , Angiografía por Resonancia Magnética , Masculino , Trombosis/fisiopatología , Ultrasonografía
9.
Oncotarget ; 7(35): 56183-56192, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27487143

RESUMEN

The transcription factor Zinc finger protein 148 (Zfp148, ZBP-89, BFCOL, BERF1, htß) interacts physically with the tumor suppressor p53, but the significance of this interaction is not known. We recently showed that knockout of Zfp148 in mice leads to ectopic activation of p53 in some tissues and cultured fibroblasts, suggesting that Zfp148 represses p53 activity. Here we hypothesize that targeting Zfp148 would unleash p53 activity and protect against cancer development, and test this idea in the APCMin/+ mouse model of intestinal adenomas. Loss of one copy of Zfp148 markedly reduced tumor numbers and tumor-associated intestinal bleedings, and improved survival. Furthermore, after activation of ß-catenin-the initiating event in colorectal cancer-Zfp148 deficiency activated p53 and induced apoptosis in intestinal explants of APCMin/+ mice. The anti-tumor effect of targeting Zfp148 depended on p53, as Zfp148 deficiency did not affect tumor numbers in APCMin/+ mice lacking one or both copies of Trp53. The results suggest that Zfp148 controls the fate of newly transformed intestinal tumor cells by repressing p53 and that targeting Zfp148 might be useful in the treatment of colorectal cancer.


Asunto(s)
Adenoma/patología , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/metabolismo , Hemorragia Gastrointestinal/patología , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenoma/mortalidad , Animales , Apoptosis , Proliferación Celular , Transformación Celular Neoplásica/patología , Células Cultivadas , Neoplasias Colorrectales/mortalidad , Proteínas de Unión al ADN/genética , Fibroblastos , Hemorragia Gastrointestinal/mortalidad , Humanos , Ratones , Ratones Noqueados , Neoplasias Experimentales/mortalidad , Neoplasias Experimentales/patología , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , beta Catenina/metabolismo
10.
Endocrinology ; 157(10): 3915-3923, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27533884

RESUMEN

Intimal hyperplasia is a vascular pathological process involved in the pathogenesis of atherosclerosis. Data suggest that T, the most important sex steroid hormone in males, protects men from atherosclerotic cardiovascular disease. T mainly acts via the androgen receptor (AR), and in this study we evaluated formation of intimal hyperplasia in male AR knockout (ARKO) mice using a vascular injury model. Two weeks after ligation of the carotid artery, male ARKO mice showed increased intimal area and intimal thickness compared with controls. After endothelial denudation by an in vivo scraping injury, there was no difference in the reendothelialization in ARKO compared with control mice. Ex vivo, we observed increased outgrowth of vascular smooth muscle cells from ARKO compared with control aortic tissue explants; the number of outgrown cells was almost doubled in ARKO. In vitro, stimulation of human aortic vascular smooth muscle cells with a physiological T concentration inhibited both migration and proliferation of the cells. Analyzing the expression of central regulators of cell proliferation and migration, we found that mRNA and protein levels of p27 were lower in uninjured arteries from ARKO mice and that T replacement to castrated male mice increased p27 mRNA in an AR-dependent manner. In conclusion, AR deficiency in male mice increases intimal hyperplasia in response to vascular injury, potentially related to the effects of androgens/AR to inhibit proliferation and migration of smooth muscle cells.


Asunto(s)
Traumatismos de las Arterias Carótidas/complicaciones , Neointima/etiología , Receptores Androgénicos/metabolismo , Testosterona/metabolismo , Animales , Traumatismos de las Arterias Carótidas/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Masculino , Ratones Noqueados , Miocitos del Músculo Liso/fisiología , Neointima/metabolismo
11.
Sci Transl Med ; 7(308): 308re8, 2015 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-26446958

RESUMEN

Antioxidants in the diet and supplements are widely used to protect against cancer, but clinical trials with antioxidants do not support this concept. Some trials show that antioxidants actually increase cancer risk and a study in mice showed that antioxidants accelerate the progression of primary lung tumors. However, little is known about the impact of antioxidant supplementation on the progression of other types of cancer, including malignant melanoma. We show that administration of N-acetylcysteine (NAC) increases lymph node metastases in an endogenous mouse model of malignant melanoma but has no impact on the number and size of primary tumors. Similarly, NAC and the soluble vitamin E analog Trolox markedly increased the migration and invasive properties of human malignant melanoma cells but did not affect their proliferation. Both antioxidants increased the ratio between reduced and oxidized glutathione in melanoma cells and in lymph node metastases, and the increased migration depended on new glutathione synthesis. Furthermore, both NAC and Trolox increased the activation of the small guanosine triphosphatase (GTPase) RHOA, and blocking downstream RHOA signaling abolished antioxidant-induced migration. These results demonstrate that antioxidants and the glutathione system play a previously unappreciated role in malignant melanoma progression.


Asunto(s)
Antioxidantes/farmacología , Melanoma/inducido químicamente , Acetilcisteína/efectos adversos , Acetilcisteína/farmacología , Animales , Antioxidantes/efectos adversos , Línea Celular Tumoral , Cromanos/efectos adversos , Cromanos/farmacología , Suplementos Dietéticos/efectos adversos , Modelos Animales de Enfermedad , Glutatión/metabolismo , Humanos , Masculino , Melanoma/patología , Ratones , Metástasis de la Neoplasia/patología
12.
Circ Res ; 115(9): 781-9, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25212213

RESUMEN

RATIONALE: Cell proliferation and cell cycle control mechanisms are thought to play central roles in the pathogenesis of atherosclerosis. The transcription factor Zinc finger protein 148 (Zfp148) was shown recently to maintain cell proliferation under oxidative conditions by suppressing p53, a checkpoint protein that arrests proliferation in response to various stressors. It is established that inactivation of p53 accelerates atherosclerosis, but whether increased p53 activation confers protection against the disease remains to be determined. OBJECTIVE: We aimed to test the hypothesis that Zfp148 deficiency reduces atherosclerosis by unleashing p53 activity. METHODS AND RESULTS: Mice harboring a gene-trap mutation in the Zfp148 locus (Zfp148(gt/+)) were bred onto the apolipoprotein E (Apoe)(-/-) genetic background and fed a high-fat or chow diet. Loss of 1 copy of Zfp148 markedly reduced atherosclerosis without affecting lipid metabolism. Bone marrow transplantation experiments revealed that the effector cell is of hematopoietic origin. Peritoneal macrophages and atherosclerotic lesions from Zfp148(gt/+)Apoe(-/-) mice showed increased levels of phosphorylated p53 compared with controls, and atherosclerotic lesions contained fewer proliferating macrophages. Zfp148(gt/+)Apoe(-/-) mice were further crossed with p53-null mice (Trp53(-/-) [the gene encoding p53]). There was no difference in atherosclerosis between Zfp148(gt/+)Apoe(-/-) mice and controls on a Trp53(+/-) genetic background, and there was no difference in levels of phosphorylated p53 or cell proliferation. CONCLUSIONS: Zfp148 deficiency increases p53 activity and protects against atherosclerosis by causing proliferation arrest of lesional macrophages, suggesting that drugs targeting macrophage proliferation may be useful in the treatment of atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Puntos de Control del Ciclo Celular , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Macrófagos Peritoneales/metabolismo , Factores de Transcripción/deficiencia , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Animales , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Trasplante de Médula Ósea , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Placa Aterosclerótica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética
13.
Sci Transl Med ; 6(221): 221ra15, 2014 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24477002

RESUMEN

Antioxidants are widely used to protect cells from damage induced by reactive oxygen species (ROS). The concept that antioxidants can help fight cancer is deeply rooted in the general population, promoted by the food supplement industry, and supported by some scientific studies. However, clinical trials have reported inconsistent results. We show that supplementing the diet with the antioxidants N-acetylcysteine (NAC) and vitamin E markedly increases tumor progression and reduces survival in mouse models of B-RAF- and K-RAS-induced lung cancer. RNA sequencing revealed that NAC and vitamin E, which are structurally unrelated, produce highly coordinated changes in tumor transcriptome profiles, dominated by reduced expression of endogenous antioxidant genes. NAC and vitamin E increase tumor cell proliferation by reducing ROS, DNA damage, and p53 expression in mouse and human lung tumor cells. Inactivation of p53 increases tumor growth to a similar degree as antioxidants and abolishes the antioxidant effect. Thus, antioxidants accelerate tumor growth by disrupting the ROS-p53 axis. Because somatic mutations in p53 occur late in tumor progression, antioxidants may accelerate the growth of early tumors or precancerous lesions in high-risk populations such as smokers and patients with chronic obstructive pulmonary disease who receive NAC to relieve mucus production.


Asunto(s)
Antioxidantes/efectos adversos , Progresión de la Enfermedad , Neoplasias Pulmonares/patología , Acetilcisteína/efectos adversos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Daño del ADN , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ratones , Especies Reactivas de Oxígeno/metabolismo , Solubilidad , Proteína p53 Supresora de Tumor/metabolismo , Vitamina E/efectos adversos , Vitamina E/análogos & derivados
14.
PLoS One ; 8(7): e70022, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936136

RESUMEN

Hematopoiesis is regulated by transcription factors that induce cell fate and differentiation in hematopoietic stem cells into fully differentiated hematopoietic cell types. The transcription factor zinc finger protein 148 (Zfp148) interacts with the hematopoietic transcription factor Gata1 and has been implicated to play an important role in primitive and definitive hematopoiesis in zebra fish and mouse chimeras. We have recently created a gene-trap knockout mouse model deficient for Zfp148, opening up for analyses of hematopoiesis in a conventional loss-of-function model in vivo. Here, we show that Zfp148-deficient neonatal and adult mice have normal or slightly increased levels of hemoglobin, hematocrit, platelets and white blood cells, compared to wild type controls. Hematopoietic lineages in bone marrow, thymus and spleen from Zfp148 (gt/gt) mice were further investigated by flow cytometry. There were no differences in T-cells (CD4 and CD8 single positive cells, CD4 and CD8 double negative/positive cells) in either organ. However, the fraction of CD69- and B220-positive cells among lymphocytes in spleen was slightly lower at postnatal day 14 in Zfp148 (gt/gt) mice compared to wild type mice. Our results demonstrate that Zfp148-deficient mice generate normal mature hematopoietic populations thus challenging earlier studies indicating that Zfp148 plays a critical role during hematopoietic development.


Asunto(s)
Médula Ósea/metabolismo , Proteínas de Unión al ADN/genética , Hematopoyesis/genética , Bazo/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Médula Ósea/embriología , Médula Ósea/crecimiento & desarrollo , Proteínas de Unión al ADN/deficiencia , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Lectinas Tipo C/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Recuento de Linfocitos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/embriología , Bazo/crecimiento & desarrollo , Timo/embriología , Timo/crecimiento & desarrollo , Factores de Tiempo , Factores de Transcripción/deficiencia
15.
PLoS One ; 8(4): e62035, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637959

RESUMEN

Genome-wide association (GWA) studies usually detect common genetic variants with low-to-medium effect sizes. Many contributing variants are not revealed, since they fail to reach significance after strong correction for multiple comparisons. The WTCCC study for hypertension, for example, failed to identify genome-wide significant associations. We hypothesized that genetic variation in genes expressed specifically in the endothelium may be important for hypertension development. Results from the WTCCC study were combined with previously published gene expression data from mice to specifically investigate SNPs located within endothelial-specific genes, bypassing the requirement for genome-wide significance. Six SNPs from the WTCCC study were selected for independent replication in 5205 hypertensive patients and 5320 population-based controls, and successively in a cohort of 16,537 individuals. A common variant (rs10860812) in the DRAM (damage-regulated autophagy modulator) locus showed association with hypertension (P = 0.008) in the replication study. The minor allele (A) had a protective effect (OR = 0.93; 95% CI 0.88-0.98 per A-allele), which replicates the association in the WTCCC GWA study. However, a second follow-up, in the larger cohort, failed to reveal an association with blood pressure. We further tested the endothelial-specific genes for co-localization with a panel of newly discovered SNPs from large meta-GWAS on hypertension or blood pressure. There was no significant overlap between those genes and hypertension or blood pressure loci. The result does not support the hypothesis that genetic variation in genes expressed in endothelium plays an important role for hypertension development. Moreover, the discordant association of rs10860812 with blood pressure in the case control study versus the larger Malmö Preventive Project-study highlights the importance of rigorous replication in multiple large independent studies.


Asunto(s)
Endotelio/metabolismo , Hipertensión/genética , Polimorfismo de Nucleótido Simple , Animales , Femenino , Sitios Genéticos/genética , Marcadores Genéticos/genética , Estudio de Asociación del Genoma Completo , Humanos , Hipertensión/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Persona de Mediana Edad , Especificidad de Órganos
16.
PLoS One ; 8(2): e55720, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23405202

RESUMEN

The transcription factor Zfp148 (Zbp-89, BFCOL, BERF1, htß) interacts physically with the tumor suppressor p53 and is implicated in cell cycle control, but the physiological role of Zfp148 remains unknown. Here we show that Zfp148 deficiency leads to respiratory distress and lethality in newborn mice. Zfp148 deficiency prevented structural maturation of the prenatal lung without affecting type II cell differentiation or surfactant production. BrdU analyses revealed that Zfp148 deficiency caused proliferation arrest of pulmonary cells at E18.5-19.5. Similarly, Zfp148-deficient fibroblasts exhibited proliferative arrest that was dependent on p53, raising the possibility that cell stress is part of the underlying mechanism. Indeed, Zfp148 deficiency lowered the threshold for activation of p53 under oxidative conditions. Moreover, both in vivo and cellular phenotypes were rescued on Trp53(+/-) or Trp53(-/-) backgrounds and by antioxidant treatment. Thus, Zfp148 prevents respiratory distress and lethality in newborn mice by attenuating oxidative stress-dependent p53-activity during the saccular stage of lung development. Our results establish Zfp148 as a novel player in mammalian lung maturation and demonstrate that Zfp148 is critical for cell cycle progression in vivo.


Asunto(s)
Antioxidantes/farmacología , Proteínas de Unión al ADN/fisiología , Eliminación de Gen , Genes Letales , Pulmón/embriología , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/genética , Animales , Animales Recién Nacidos , Apoptosis , Southern Blotting , Western Blotting , Ciclo Celular , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnicas para Inmunoenzimas , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Enfermedades Respiratorias/genética , Enfermedades Respiratorias/patología , Enfermedades Respiratorias/prevención & control , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/deficiencia
17.
Nature ; 464(7290): 917-21, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20228789

RESUMEN

The vascular endothelial growth factors (VEGFs) are major angiogenic regulators and are involved in several aspects of endothelial cell physiology. However, the detailed role of VEGF-B in blood vessel function has remained unclear. Here we show that VEGF-B has an unexpected role in endothelial targeting of lipids to peripheral tissues. Dietary lipids present in circulation have to be transported through the vascular endothelium to be metabolized by tissue cells, a mechanism that is poorly understood. Bioinformatic analysis showed that Vegfb was tightly co-expressed with nuclear-encoded mitochondrial genes across a large variety of physiological conditions in mice, pointing to a role for VEGF-B in metabolism. VEGF-B specifically controlled endothelial uptake of fatty acids via transcriptional regulation of vascular fatty acid transport proteins. As a consequence, Vegfb(-/-) mice showed less uptake and accumulation of lipids in muscle, heart and brown adipose tissue, and instead shunted lipids to white adipose tissue. This regulation was mediated by VEGF receptor 1 and neuropilin 1 expressed by the endothelium. The co-expression of VEGF-B and mitochondrial proteins introduces a novel regulatory mechanism, whereby endothelial lipid uptake and mitochondrial lipid use are tightly coordinated. The involvement of VEGF-B in lipid uptake may open up the possibility for novel strategies to modulate pathological lipid accumulation in diabetes, obesity and cardiovascular diseases.


Asunto(s)
Endotelio/metabolismo , Ácidos Grasos/metabolismo , Factor B de Crecimiento Endotelial Vascular/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Transporte Biológico , Línea Celular , Núcleo Celular/genética , Células Cultivadas , Endotelio/citología , Proteínas de Transporte de Ácidos Grasos/genética , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Músculos/metabolismo , Miocardio/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Transducción de Señal , Transcripción Genética , Factor B de Crecimiento Endotelial Vascular/deficiencia , Factor B de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Genome Med ; 1(11): 108, 2009 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-19917099

RESUMEN

BACKGROUND: A function for the microRNA (miRNA) pathway in vascular development and angiogenesis has been firmly established. miRNAs with selective expression in the vasculature are attractive as possible targets in miRNA-based therapies. However, little is known about the expression of miRNAs in microvessels in vivo. Here, we identified candidate microvascular-selective miRNAs by screening public miRNA expression datasets. METHODS: Bioinformatics predictions of microvascular-selective expression were validated with real-time quantitative reverse transcription PCR on purified microvascular fragments from mouse. Pericyte expression was shown with in situ hybridization on tissue sections. Target sites were identified with 3' UTR luciferase assays, and migration was tested in a microfluid chemotaxis chamber. RESULTS: miR-145, miR-126, miR-24, and miR-23a were selectively expressed in microvascular fragments isolated from a range of tissues. In situ hybridization and analysis of Pdgfb retention motif mutant mice demonstrated predominant expression of miR-145 in pericytes. We identified the Ets transcription factor Friend leukemia virus integration 1 (Fli1) as a miR-145 target, and showed that elevated levels of miR-145 reduced migration of microvascular cells in response to growth factor gradients in vitro. CONCLUSIONS: miR-126, miR-24 and miR-23a are selectively expressed in microvascular endothelial cells in vivo, whereas miR-145 is expressed in pericytes. miR-145 targets the hematopoietic transcription factor Fli1 and blocks migration in response to growth factor gradients. Our findings have implications for vascular disease and provide necessary information for future drug design against miRNAs with selective expression in the microvasculature.

19.
FASEB J ; 23(5): 1490-502, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19136612

RESUMEN

To define molecular events accompanying formation of the 3-dimensional (3D) vascular tube, we have characterized gene expression during vascular endothelial growth factor (VEGF)-induced tubular morphogenesis of endothelial cells. Microarray analyses were performed comparing gene induction in growth-arrested, tube-forming endothelial cells harvested from 3D collagen cultures to that in proliferating endothelial cells cultured on fibronectin. Differentially expressed genes were clustered and analyzed for specific endothelial expression through publicly available datasets. We validated the contribution of one of the identified genes, vascular endothelial protein tyrosine phosphatase (VE-PTP), to endothelial morphogenesis. Silencing of VE-PTP expression was accompanied by increased VEGF receptor-2 (VEGFR2) tyrosine phosphorylation and activation of downstream signaling pathways. The increased VEGFR2 activity promoted endothelial cell cycle progression, overcoming the G(0)/G(1) arrest associated with organization into tubular structures in the 3D cultures. Proximity ligation showed close association between VEGFR2 and VE-PTP in resting cells. Activation of VEGFR2 by VEGF led to rapid loss of association, which was resumed with time in parallel with decreased receptor activity. In conclusion, we have identified genes, which may serve critical functions in formation of the vascular tube. One of these, VE-PTP, regulates VEGFR2 activity thereby modulating the VEGF-response during angiogenesis.


Asunto(s)
Células Endoteliales/ultraestructura , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Células Cultivadas , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Morfogénesis/genética , Transducción de Señal
20.
Circ Res ; 103(1): 61-9, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18511849

RESUMEN

Lipoma preferred partner (LPP) was recently recognized as a smooth muscle marker that plays a role in smooth muscle cell migration. In this report, we focus on the transcriptional regulation of the LPP gene. In particular, we investigate whether LPP is directly regulated by serum response factor (SRF). We show that the LPP gene contains 3 evolutionarily conserved CArG boxes and that 1 of these is part of an alternative promoter in intron 2. Quantitative RT-PCR shows that this alternative promoter directs transcription specifically to smooth muscle containing tissues in vivo. By using chromatin immunoprecipitation, we demonstrate that 2 of the CArG boxes, including the promoter-associated CArG box, bind to endogenous SRF in cultured aortic smooth muscle cells. Electrophoretic mobility-shift assays show that the conserved CArG boxes bind SRF in vitro. In reporter experiments, we show that the alternative promoter has transcriptional capacity that is dependent on SRF/myocardin and that the promoter associated CArG box is required for that activity. Finally, we show by quantitative RT-PCR that the alternative promoter is strongly downregulated in SRF-deficient embryonic stem cells and in smooth muscle tissues derived from conditional SRF knockout mice. Collectively, our data demonstrate that expression of LPP in smooth muscle is mediated by an alternative promoter that is regulated by SRF/myocardin.


Asunto(s)
Aorta/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Intrones/fisiología , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Elemento de Respuesta al Suero/fisiología , Factor de Respuesta Sérica/metabolismo , Transactivadores/metabolismo , Animales , Aorta/citología , Movimiento Celular/fisiología , Células Cultivadas , Proteínas del Citoesqueleto/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas con Dominio LIM , Masculino , Ratones , Miocitos del Músculo Liso/citología , Proteínas Nucleares/genética , Factor de Respuesta Sérica/genética , Transactivadores/genética , Transcripción Genética/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA