Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Oncogene ; 40(34): 5275-5285, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34244607

RESUMEN

Endometrial carcinoma (EC) is the fourth-most common cancer in women in the United States, and generally carries a favorable prognosis. However, about 10% of EC patients have a rare and aggressive form, uterine serous papillary carcinoma (USPC), which carries a much higher mortality rate. The developmental transcription factor PAX8 is expressed in nearly 100% of USPCs. We show that PAX8 plays a critical antiapoptotic role in USPC and this role is established via transcriptional activation of two aberrant signaling pathways. First, PAX8 positively regulates mutated p53, and missense p53 mutations have an oncogenic gain of function effect. Second, PAX8 directly transcriptionally regulates p21, in a p53-independent manner, and p21 acquires a growth promoting role that is mediated via cytoplasmic localization of the protein. We propose that mutated p53 and cytoplasmic p21 can independently mediate the pro-proliferative role of PAX8 in USPC. In addition, we performed a genome-wide transcriptome analysis to detect pathways that are regulated by PAX8, and propose that metabolism and HIF-1alpha -related pathways are potential candidates for mediating the role of PAX8 in USPC. Taken together our findings demonstrate for the first time that PAX8 is an essential lineage marker in USPC, and suggest its mechanism of action.


Asunto(s)
Cistadenocarcinoma Seroso , Oncogenes , Apoptosis , Neoplasias Endometriales , Femenino , Perfilación de la Expresión Génica , Humanos
2.
Am J Respir Cell Mol Biol ; 65(2): 214-221, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33891828

RESUMEN

Obesity elevates the plasma level of leptin, which has been associated with hypertension. Our recent studies in mice demonstrated that leptin increases blood pressure by activating the carotid sinus nerve, which transmits the chemosensory input from carotid bodies (CBs) to the medullary centers, and that the effect of leptin is mediated via Trpm7 (TRP [transient receptor potential] melastatin 7) channels in CB glomus cells. We also found that Trpm7 overexpression and Trpm7 promoter demethylation in CBs correlate positively with the hyperleptinemia and leptin receptor overexpression in CBs. Hence, we postulated that leptin epigenetically regulates Trpm7 expression in CBs. We addressed our hypothesis by using rat adrenal pheochromocytoma (PC12) cells as a model of CB glomus cells. PC12 cells expressing LEPRb (long, active form of leptin receptor) showed dramatic induction of the promoter activity and expression of Trpm7 upon leptin treatment. The increased Trpm7 expression coincided with the reduction of CpG site-specific methylation and trimethylation of H3K27 (H3 [histone 3] K27 [lysine 27]) and the increase of acetylation of H3K27 and trimethylation of H3K4 (H3 lysine 4) at the Trpm7 promoter. The inhibitor of STAT3 (signal transducer and activator of transcription 3) signaling, SD1008, reversed the leptin-induced Trpm7 promoter activity via modulations of the binding of pSTAT3 (phosphorylated STAT3) and DNMT3B (DNA methyltransferase 3B) and modifications of H3K27 and H3K4 at the Trpm7 promoter. Our results suggest that leptin-activated pSTAT3 epigenetically regulates the transcription of Trpm7 through DNA methylation and histone modifications. Because epigenetic changes are reversible, targeting epigenetic modifications of Trpm7 may serve as a new therapeutic approach for the treatment of hypertension in obesity.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/metabolismo , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Leptina/farmacología , Proteínas de Neoplasias/biosíntesis , Feocromocitoma/metabolismo , Canales Catiónicos TRPM/biosíntesis , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/patología , Animales , Proteínas de Neoplasias/genética , Células PC12 , Feocromocitoma/genética , Feocromocitoma/patología , Ratas , Canales Catiónicos TRPM/genética
3.
Nanomedicine ; 14(8): 2656-2665, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30010000

RESUMEN

The 3-D spatial and mechanical features of nano-topography can create alternative environments, which influence cellular response. In this paper, murine fibroblast cells were grown on surfaces characterized by protruding nanotubes. Cells cultured on such nano-structured surface exhibit stronger cellular adhesion compared to control groups, but despite the fact that stronger adhesion is generally believed to promote cell cycle progression, the time cells spend in G1 phase is doubled. This apparent contradiction is solved by confocal microscopy analysis, which shows that the nano-topography inhibits actin stress fiber formation. In turn, this impairs RhoA activation, which is required to suppress the inhibition of cell cycle progression imposed by p21/p27. This finding suggests that the generation of stress fibers, required to impose the homeostatic intracellular tension, rather than cell adhesion/spreading is the limiting factor for cell cycle progression. Indeed, nano-topography could represent a unique tool to inhibit proliferation in adherent well-spread cells.


Asunto(s)
Adhesión Celular , Ciclo Celular , Fibroblastos/fisiología , Nanoestructuras/química , Animales , División Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibroblastos/citología , Ratones , Andamios del Tejido , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
4.
Eur J Pharm Biopharm ; 114: 1-10, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28088004

RESUMEN

We validated the anticancer potential of a nanoformulation made by (+)-catechin, gelatin and carbon nanotubes in terms of inhibition of cancer cell proliferation, migration and associated neo-angiogenesis. Gelatin was selected to stabilize the catechin without compromising its anti-oxidant potential and the carbon nanotubes were used to increase its intracellular bioavailability. The anticancer potential of the resulting nanohybrid was validated on an aggressive melanoma cell line, in vitro and in zebrafish xenotransplants. The nanohybrid strongly enhances the cytotoxic effect of (+)-catechin. At a concentration of (+)-catechin 50µg/ml, the nanohybrid inhibited the ability of melanoma cells to proliferate (100% increase of cell doubling time and severe impairment in zebrafish xenotransplants), to migrate (totally inhibition in vitro and 50% reduction of cell motility in zebrafish xenotransplants) and to induce neo-angiogenesis (100% inhibition in zebrafish xenotransplants). Conversely, the free (+)-catechin and carrier (CNT:gel) had no statistically significant effects over the control, at any concentration tested. Our results suggest that the use of the nanohybrid, able to improve the therapeutic efficacy of the catechins, could represent a successful strategy for a future clinical translation.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Catequina/química , Catequina/farmacología , Proliferación Celular/efectos de los fármacos , Melanoma/tratamiento farmacológico , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Catequina/administración & dosificación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Composición de Medicamentos , Humanos , Melanoma/patología , Nanopartículas , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
5.
Stem Cells ; 34(4): 1011-26, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26676563

RESUMEN

Non-neoplastic stromal cells harvested from patient tumors were identified as tumor-derived mesenchymal stem cells (MSCs) by their multipotential capacity to differentiate into adipocytes, osteoblasts, and chondrocytes and by the expression of MSC specific cell surface markers. These procedures yielded also epithelial cancer cells and their counterpart MSC from gastric carcinoma (GSC1) and lung carcinoma (LC2). While the LC2 cancer cell growth is independent of their LC-MSC, the GSC1 cancer cell growth is critically dependent on the presence of their counterpart GSC-MSC or their conditioned medium (CM). The fact that none of the various other tumor-derived MSCs was able to restore the specific effect of GSC-MSC on GSC1 cancer cell growth suggests specificity of tumor-derived MSC, which are specifically recruited and "educated"/reprogrammed by the cancer cells to support tumor growth. Using cytokine array analysis, we were able to demonstrate that GSC1 cell growth is mediated through hepatocyte growth factor (HGF)/c-MET signaling pathway which is activated exclusively by HGF secreted from GSC-MSC. An innovative approach demonstrates GSC1-mediated specific tropism of "naïve" MSC from the adjacent tissue in a tumor specific manner to support tumor progression. The results suggest that specific tumor tropic "naïve" MSC are reprogrammed in a tumor-specific manner to support gastric tumor progression. Understanding the mechanisms involved in the interactions of the tumor cancer cells and tumor-derived MSC will constitute the basis for developing multimodal anticancer therapeutic strategies that will also take into account the specific tumor tropism properties of MSC and their reprogramming.


Asunto(s)
Carcinoma/patología , Factor de Crecimiento de Hepatocito/biosíntesis , Neoplasias Pulmonares/patología , Células Madre Mesenquimatosas/patología , Proteínas Proto-Oncogénicas c-met/biosíntesis , Neoplasias Gástricas/patología , Adipocitos/metabolismo , Carcinogénesis/genética , Carcinoma/genética , Carcinoma/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Condrocitos/metabolismo , Medios de Cultivo Condicionados/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Osteoblastos/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
6.
Hum Genet ; 134(3): 305-14, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25560765

RESUMEN

Lissencephaly comprises a heterogeneous group of developmental brain disorders of varying severity, involving abnormal cortical gyration. We studied a highly consanguineous Israeli Moslem family with a lethal form of autosomal recessive lissencephaly with cerebellar hypoplasia (LCH). Using microarray-based homozygosity mapping in the reported family, combined with whole exome sequencing in one affected infant, we identified a homozygous splice site mutation g.IVS8+1G>A in cyclin-dependent kinase 5 (CDK5), causing complete skipping of exon 8, and leading to a frame shift and premature stop codon (p.V162SfsX19). The mutation co-segregated with the disease phenotype in all 29 study participants (4 patients and 25 healthy relatives), and was not identified in 200 ethnically matched control chromosomes. The p.V162SfsX19 mutation causes lack of endogenous CDK5 expression in affected dermal fibroblasts and brain tissue at the mRNA and protein levels, consistent with nonsense-mediated mRNA decay. Functional analysis of the p.V162SfsX19 mutation, using a yeast complementation assay, showed loss-of-function of the mutant CDK5 gene product, thereby implicating its role in the pathogenesis of autosomal recessive LCH in the studied family.


Asunto(s)
Cerebelo/anomalías , Quinasa 5 Dependiente de la Ciclina/genética , Lisencefalia/genética , Malformaciones del Sistema Nervioso/genética , Secuencia de Bases , Células Cultivadas , Cerebelo/enzimología , Consanguinidad , Análisis Mutacional de ADN , Discapacidades del Desarrollo/enzimología , Discapacidades del Desarrollo/genética , Femenino , Genes Recesivos , Estudios de Asociación Genética , Prueba de Complementación Genética , Homocigoto , Humanos , Lactante , Recién Nacido , Lisencefalia/enzimología , Masculino , Mutación Missense , Malformaciones del Sistema Nervioso/enzimología , Linaje
8.
PLoS One ; 8(12): e83651, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358304

RESUMEN

Intratumoral heterogeneity challenges existing paradigms for anti-cancer therapy. We have previously demonstrated that the human embryonic stem cells (hESC)-derived cellular microenvironment in immunocompromised mice, enables functional distinction of heterogeneous tumor cells, including cells which do not grow into a tumor in a conventional direct tumor xenograft platform. We have identified and characterized six cancer cell subpopulations each clonally expanded from a single cell, derived from human ovarian clear cell carcinoma of a single tumor, to demonstrate striking intratumoral phenotypic heterogeneity that is dynamically dependent on the tumor growth microenvironment. These cancer cell subpopulations, characterized as cancer stem cell subpopulations, faithfully recapitulate the full spectrum of histological phenotypic heterogeneity known for human ovarian clear cell carcinoma. Each of the six subpopulations displays a different level of morphologic and tumorigenic differentiation wherein growth in the hESC-derived microenvironment favors growth of CD44+/aldehyde dehydrogenase positive pockets of self-renewing cells that sustain tumor growth through a process of tumorigenic differentiation into CD44-/aldehyde dehydrogenase negative derivatives. Strikingly, these derivative cells display microenvironment-dependent plasticity with the capacity to restore self-renewal markers and CD44 expression. In the current study, we delineate the distinct gene expression and epigenetic profiles of two such subpopulations, representing extremes of phenotypic heterogeneity in terms of niche-dependent self-renewal and tumorigenic differentiation. By combining Gene Set Enrichment, Gene Ontology and Pathway-focused array analyses with methylation status, we propose a suite of robust differences in tumor self-renewal and differentiation pathways that underlie the striking intratumoral phenotypic heterogeneity which characterize this and other solid tumor malignancies.


Asunto(s)
Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Nicho de Células Madre/genética , Animales , Células Cultivadas , Femenino , Humanos , Ratones , Ratones SCID , Análisis por Micromatrices , Persona de Mediana Edad , Transcriptoma
9.
Stem Cells ; 30(3): 415-24, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22267284

RESUMEN

Resistance to anticancer therapy has been attributed to interindividual differences in gene expression pathways among tumors, and to the existence within tumors of cancer stem cells with self-renewal capacity. In previous studies, we have demonstrated that the human embryonic stem cell (hESC)-derived cellular microenvironment in immunocompromised mice enables functional distinction of heterogeneous tumor cells, including cells that do not grow into a tumor in conventional direct tumor xenograft platform. In the current study, we use clonally expanded subpopulations derived from ovarian clear cell carcinoma of a single tumor, to demonstrate striking intratumoral phenotypic heterogeneity that is dynamically dependent on the tumor growth microenvironment. Each of six clonally expanded subpopulations displays a different level of morphologic and tumorigenic differentiation, wherein growth in the hESC-derived microenvironment favors growth of CD44+ aldehyde dehydrogenase positive pockets of self-renewing cells that sustain tumor growth through a process of tumorigenic differentiation into CD44- aldehyde dehydrogenase negative derivatives. Strikingly, these derivative cells display microenvironment-dependent plasticity with the capacity to restore self-renewal and CD44 expression. Such intratumoral heterogeneity and plasticity at the level of the key properties of self-renewal and tumorigenic differentiation suggests that a paradigm shift is needed in the approach to anticancer therapy, with the aim of turning malignant growth into a chronic manageable disorder, based on continual monitoring of these tumor growth properties. The hESC-based in vivo model renders intratumoral heterogeneity in the self-renewal and tumorigenic differentiation amenable to biological analysis as well as anticancer therapy testing.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Receptores de Hialuranos/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/patología , Animales , Técnicas de Cultivo de Célula , Transformación Celular Neoplásica , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Nicho de Células Madre , Células Tumorales Cultivadas
10.
Endocrine ; 41(1): 53-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22169961

RESUMEN

Gitelman's syndrome (GS), also known as familial hypokalemic hypomagnesemia, is a rare autosomal recessive hereditary salt-losing tubulopathy, characterized by hypokalemic metabolic alkalosis, hypomagnesemia, and hypocalciuria, which is usually caused by mutations in the SLC12A3 gene encoding the thiazide-sensitive sodium chloride contrasporter. Because 18-40% of suspected GS patients carry only one SLC12A3 mutant allele, large genomic rearrangements must account for unidentified mutations. The clinical manifestations of GS are highly variable in terms of age at presentation, severity of symptoms, and biochemical abnormalities. Molecular analysis in our sibling's patients revealed compound heterozygous mutations in the coding region of SLC12A3 as underlying their disease. Such compound heterozygosity can result in disease phenotype for such loss of function mutations in the absence of homozygosis through consanguineous inheritance of mutant alleles, identical by descent. Missense mutations account for approximately 70% of the mutations in GS, and there is a predisposition to large rearrangements caused by the presence of repeated sequences within the SLC12A3. We report two adult male siblings of Jewish origin with late onset GS, who presented in their fifth decade of life with muscle weakness, hypokalemia, hypomagnesaemia, and metabolic alkalosis. Rapid clinical and biochemical improvement was achieved by replacement therapy with potassium and magnesium.


Asunto(s)
Síndrome de Gitelman/genética , Síndrome de Gitelman/fisiopatología , Mutación/genética , Receptores de Droga/genética , Simportadores/genética , Adulto , Secuencia de Aminoácidos , Animales , Síndrome de Gitelman/tratamiento farmacológico , Humanos , Magnesio/uso terapéutico , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Potasio/uso terapéutico , Miembro 3 de la Familia de Transportadores de Soluto 12 , Resultado del Tratamiento
11.
N Engl J Med ; 362(12): 1102-9, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20335586

RESUMEN

We describe two siblings from a consanguineous family with autosomal recessive Fanconi's syndrome and hypophosphatemic rickets. Genetic analysis revealed a homozygous in-frame duplication of 21 bp in SLC34A1, which encodes the renal sodium-inorganic phosphate cotransporter NaPi-IIa, as the causative mutation. Functional studies in Xenopus laevis oocytes and in opossum kidney cells indicated complete loss of function of the mutant NaPi-IIa, resulting from failure of the transporter to reach the plasma membrane. These findings show that disruption of the human NaPi-IIa profoundly impairs overall renal phosphate reabsorption and proximal-tubule function and provide evidence of the critical role of NaPi-IIa in human renal phosphate handling.


Asunto(s)
Raquitismo Hipofosfatémico Familiar/genética , Síndrome de Fanconi/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Adulto , Animales , Calcitriol/sangre , Células Cultivadas , Consanguinidad , Análisis Mutacional de ADN , Femenino , Genes Recesivos , Humanos , Riñón/citología , Riñón/metabolismo , Masculino , Mutación , Oocitos/metabolismo , Zarigüeyas , Linaje , Hermanos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...