Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Dev Pathol ; 26(4): 394-403, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37334553

RESUMEN

BACKGROUND: Hepatocellular adenoma (HCA) in the pediatric population is very rare and there are only limited studies, especially with molecular characterization of the tumors. Main HCA subtypes recognized in the current WHO classification include HNF1A-inactivated HCA (H-HCA), inflammatory HCA (IHCA), ß-catenin-activated HCA (b-HCA), and ß-catenin-activated IHCA (b-IHCA) and sonic hedgehog HCA (shHCA) is reported as an emerging subtype. METHODS: Clinical history, pathological information, and molecular studies for a series of 2 cases of pediatric HCA were reviewed. RESULTS: Case 1 was a b-HCA characterized by somatic CTNNB1 S45 mutation in a 11-year-old male with Abernethy malformation. Case 2 was a H-HCA characterized by germline HNF1A variant (c.526+1G>A) in a 15-year-old male associated with maturity-onset diabetes of the young type 3 (MODY3). CONCLUSION: Our findings highlight the rarity of these 2 cases associated with adenomatosis, and the contribution of molecular/genetic analysis for proper sub-typing, prognosis and family surveillance.


Asunto(s)
Adenoma de Células Hepáticas , Carcinoma Hepatocelular , Neoplasias Hepáticas , Masculino , Humanos , Niño , Adolescente , Adenoma de Células Hepáticas/diagnóstico , Adenoma de Células Hepáticas/genética , Adenoma de Células Hepáticas/patología , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , beta Catenina/genética , Proteínas Hedgehog , Fenotipo , Genotipo
2.
Blood ; 136(23): 2691-2702, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-32659785

RESUMEN

The mechanisms by which phlebotomy promotes the mobilization of hepatic iron stores are not well understood. NCOA4 (nuclear receptor coactivator 4) is a widely expressed intracellular protein previously shown to mediate the autophagic degradation of ferritin. Here, we investigate a local requirement for NCOA4 in the regulation of hepatic iron stores and examine mechanisms of NCOA4 regulation. Hepatocyte-targeted Ncoa4 knockdown in nonphlebotomized mice had only modest effects on hepatic ferritin subunit levels and nonheme iron concentration. After phlebotomy, mice with hepatocyte-targeted Ncoa4 knockdown exhibited anemia and hypoferremia similar to control mice with intact Ncoa4 regulation but showed a markedly impaired ability to lower hepatic ferritin subunit levels and hepatic nonheme iron concentration. This impaired hepatic response was observed even when dietary iron was limited. In both human and murine hepatoma cell lines, treatment with chemicals that stabilize hypoxia inducible factor (HIF), including desferrioxamine, cobalt chloride, and dimethyloxalylglycine, raised NCOA4 messenger RNA. This NCOA4 messenger RNA induction occurred within 3 hours, preceded a rise in NCOA4 protein, and was attenuated in the setting of dual HIF-1α and HIF-2α knockdown. In summary, we show for the first time that NCOA4 plays a local role in facilitating iron mobilization from the liver after blood loss and that HIF regulates NCOA4 expression in cells of hepatic origin. Because the prolyl hydroxylases that regulate HIF stability are oxygen- and iron-dependent enzymes, our findings suggest a novel mechanism by which hypoxia and iron deficiency may modulate NCOA4 expression to impact iron homeostasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hemorragia/metabolismo , Hepatocitos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hierro/metabolismo , Hígado/metabolismo , Coactivadores de Receptor Nuclear/biosíntesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hemorragia/genética , Hemorragia/patología , Hepatocitos/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hígado/patología , Ratones , Coactivadores de Receptor Nuclear/genética
3.
Int J Gynecol Pathol ; 41(3): 258-267, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33990091

RESUMEN

Patients with germline TP53 mutations are characterized by the occurrence of multiple early-onset malignancies. The characteristic syndrome is Li-Fraumeni syndrome (OMIM # 151623), an autosomal dominant disorder typified by premenopausal breast carcinoma, adrenal cortical tumors, bone and soft tissue sarcomas, leukemias, and tumors of the brain and spinal cord. Gynecologic malignancies are uncommonly reported in families harboring TP53 mutations, and the predominant tumor type reported is ovarian. Uterine carcinoma has been reported only a handful of times in patients with germline TP53 mutations, none as a presenting tumor in a teenager. We report on an 18-year-old patient who presented with grade 3, high-stage endometrioid endometrial carcinoma. Sequencing detected a single-nucleotide substitution in the TP53 gene (NM_000546.6:c.818G>A), encoding the missense substitution p.Arg273His (R273H) in both the tumor and normal tissue, consistent with a germline mutation. We discuss the biology of the TP53 gene and p53 protein, with emphasis on the R273H mutation. We also review the literature on endometrial carcinoma in patients with germline TP53 mutations.


Asunto(s)
Neoplasias Endometriales , Síndrome de Li-Fraumeni , Adolescente , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/genética , Femenino , Genes p53/genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Síndrome de Li-Fraumeni/complicaciones , Síndrome de Li-Fraumeni/genética , Proteína p53 Supresora de Tumor/genética
4.
Blood ; 134(18): 1547-1557, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31439541

RESUMEN

The mechanisms underlying thrombocytosis in patients with iron deficiency anemia remain unknown. Here, we present findings that support the hypothesis that low iron biases the commitment of megakaryocytic (Mk)-erythroid progenitors (MEPs) toward the Mk lineage in both human and mouse. In MEPs of transmembrane serine protease 6 knockout (Tmprss6-/-) mice, which exhibit iron deficiency anemia and thrombocytosis, we observed a Mk bias, decreased labile iron, and decreased proliferation relative to wild-type (WT) MEPs. Bone marrow transplantation assays suggest that systemic iron deficiency, rather than a local role for Tmprss6-/- in hematopoietic cells, contributes to the MEP lineage commitment bias observed in Tmprss6-/- mice. Nontransgenic mice with acquired iron deficiency anemia also show thrombocytosis and Mk-biased MEPs. Gene expression analysis reveals that messenger RNAs encoding genes involved in metabolic, vascular endothelial growth factor, and extracellular signal-regulated kinase (ERK) pathways are enriched in Tmprss6-/- vs WT MEPs. Corroborating our findings from the murine models of iron deficiency anemia, primary human MEPs exhibit decreased proliferation and Mk-biased commitment after knockdown of transferrin receptor 2, a putative iron sensor. Signal transduction analyses reveal that both human and murine MEP have lower levels of phospho-ERK1/2 in iron-deficient conditions compared with controls. These data are consistent with a model in which low iron in the marrow environment affects MEP metabolism, attenuates ERK signaling, slows proliferation, and biases MEPs toward Mk lineage commitment.


Asunto(s)
Anemia Ferropénica/metabolismo , Diferenciación Celular/fisiología , Células Progenitoras de Megacariocitos/metabolismo , Megacariocitos/metabolismo , Anemia Ferropénica/complicaciones , Animales , Proliferación Celular , Humanos , Hierro , Células Progenitoras de Megacariocitos/citología , Megacariocitos/citología , Ratones , Ratones Noqueados , Trombocitosis/etiología , Trombocitosis/metabolismo
5.
Blood ; 139(16): 2424-2425, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35446376
10.
Blood ; 134(17): 1363-1364, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31698431
11.
Haematologica ; 106(2): 326-328, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522782
15.
Curr Opin Hematol ; 20(3): 208-14, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23426198

RESUMEN

PURPOSE OF REVIEW: The circulating peptide hepcidin modulates systemic iron balance by limiting the absorption of dietary iron and the release of iron from macrophage stores. Recent studies conducted in humans, animal models, and tissue culture systems have enhanced our understanding of the molecular mechanisms by which hepcidin levels are altered in response to iron stores, inflammation, and erythropoietic activity. RECENT FINDINGS: The bone morphogenetic protein (BMP) type I receptors ALK2 and ALK3 play key, nonredundant roles in mediating hepcidin synthesis through the BMP signaling pathway. Actions of the hereditary hemochromatosis proteins HFE and transferrin receptor 2 may intersect with the BMP pathway. Hepcidin induction in response to inflammation requires cooperative BMP signaling. A variety of innate immune and infectious stimuli induce hepcidin expression. The hypoxia inducible factor pathway appears to suppress hepcidin indirectly through the capacity of erythropoietin to stimulate erythropoiesis. SUMMARY: Study of the molecular mechanisms underlying the regulation of hepcidin synthesis has revealed complex biology. Improved understanding of the signaling pathways involved in hepcidin regulation may contribute to improved therapeutic outcomes for patients with genetic and acquired disorders that impact systemic iron balance.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Homeostasis/fisiología , Trastornos del Metabolismo del Hierro/fisiopatología , Hierro de la Dieta/metabolismo , Hierro/metabolismo , Eritropoyesis/fisiología , Hepcidinas , Humanos , Transducción de Señal/fisiología
16.
Am J Physiol Renal Physiol ; 304(5): F522-32, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23269648

RESUMEN

Unlike human patients with mutations in the 56-kDa B1 subunit isoform of the vacuolar proton-pumping ATPase (V-ATPase), B1-deficient mice (Atp6v1b1(-/-)) do not develop metabolic acidosis under baseline conditions. This is due to the insertion of V-ATPases containing the alternative B2 subunit isoform into the apical membrane of renal medullary collecting duct intercalated cells (ICs). We previously reported that quantitative Western blots (WBs) from whole kidneys showed similar B2 protein levels in Atp6v1b1(-/-) and wild-type mice (Paunescu TG, Russo LM, Da Silva N, Kovacikova J, Mohebbi N, Van Hoek AN, McKee M, Wagner CA, Breton S, Brown D. Am J Physiol Renal Physiol 293: F1915-F1926, 2007). However, WBs from renal medulla (including outer and inner medulla) membrane and cytosol fractions reveal a decrease in the levels of the ubiquitous V-ATPase E1 subunit. To compare V-ATPase expression specifically in ICs from wild-type and Atp6v1b1(-/-) mice, we crossed mice in which EGFP expression is driven by the B1 subunit promoter (EGFP-B1(+/+) mice) with Atp6v1b1(-/-) mice to generate novel EGFP-B1(-/-) mice. We isolated pure IC populations by fluorescence-assisted cell sorting from EGFP-B1(+/+) and EGFP-B1(-/-) mice to compare their V-ATPase subunit protein levels. We report that V-ATPase A, E1, and H subunits are all significantly downregulated in EGFP-B1(-/-) mice, while the B2 protein level is considerably increased in these animals. We conclude that under baseline conditions B2 upregulation compensates for the lack of B1 and is sufficient to maintain basal acid-base homeostasis, even when other V-ATPase subunits are downregulated.


Asunto(s)
Células Epiteliales/metabolismo , Riñón/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Animales , Membrana Celular/metabolismo , Células Epiteliales/citología , Citometría de Flujo , Riñón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Regulación hacia Arriba , ATPasas de Translocación de Protón Vacuolares/metabolismo
17.
Blood ; 117(17): 4590-9, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21355094

RESUMEN

The hereditary hemochromatosis protein HFE promotes the expression of hepcidin, a circulating hormone produced by the liver that inhibits dietary iron absorption and macrophage iron release. HFE mutations are associated with impaired hepatic bone morphogenetic protein (BMP)/SMAD signaling for hepcidin production. TMPRSS6, a transmembrane serine protease mutated in iron-refractory iron deficiency anemia, inhibits hepcidin expression by dampening BMP/SMAD signaling. In the present study, we used genetic approaches in mice to examine the relationship between Hfe and Tmprss6 in the regulation of systemic iron homeostasis. Heterozygous loss of Tmprss6 in Hfe(-/-) mice reduced systemic iron overload, whereas homozygous loss caused systemic iron deficiency and elevated hepatic expression of hepcidin and other Bmp/Smad target genes. In contrast, neither genetic loss of Hfe nor hepatic Hfe overexpression modulated the hepcidin elevation and systemic iron deficiency of Tmprss6(-/-) mice. These results indicate that genetic loss of Tmprss6 increases Bmp/Smad signaling in an Hfe-independent manner that can restore Bmp/Smad signaling in Hfe(-/-) mice. Furthermore, these results suggest that natural genetic variation in the human ortholog TMPRSS6 might modify the clinical penetrance of HFE-associated hereditary hemochromatosis, raising the possibility that pharmacologic inhibition of TMPRSS6 could attenuate iron loading in this disorder.


Asunto(s)
Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Hierro/metabolismo , Proteínas de la Membrana/genética , Serina Endopeptidasas/genética , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Femenino , Genotipo , Hemocromatosis/genética , Hemocromatosis/metabolismo , Hemocromatosis/fisiopatología , Proteína de la Hemocromatosis , Hepcidinas , Heterocigoto , Antígenos de Histocompatibilidad Clase I/metabolismo , Homocigoto , Humanos , Hígado/fisiología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Serina Endopeptidasas/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
18.
Am J Dermatopathol ; 35(3): 351-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23051628

RESUMEN

OBJECTIVE: To test the hypothesis that the subset of patients with impaired renal function who are exposed to gadolinium-containing contrast agents (GCCAs) and develop nephrogenic systemic fibrosis (NSF) have a genetic predisposition for disease. METHODS: We examined whether an intronic single-nucleotide polymorphism (SNP) in caveolin-1 (CAV1 rs4730751) and 2 coding SNPs in transforming growth factor-beta 1 (TGFB1 rs1800471, codon 25; and rs1800470, codon 10) were associated with the NSF phenotype. RESULTS: Forty-one patients with a history of chronic kidney disease and GCCA administration were studied, including NSF cases (n = 17) and control subjects (n = 24) without clinical or histological evidence of NSF. No significant differences in the genotype frequencies at these SNPs in TGFB1 and CAV1 were found between patients with NSF and subjects without NSF. CONCLUSIONS: We conclude that polymorphisms in the genes encoding TGFB1 and CAV1 previously associated with the development and progression of fibrosis in several organ systems are not associated with development of NSF in this cohort of patients with renal impairment after GCCA exposure.


Asunto(s)
Caveolina 1/genética , Dermopatía Fibrosante Nefrogénica/genética , Polimorfismo de Nucleótido Simple , Factor de Crecimiento Transformador beta1/genética , Adulto , Anciano , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Codón , Medios de Contraste/efectos adversos , Femenino , Fibrosis , Gadolinio/efectos adversos , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Intrones , Masculino , Persona de Mediana Edad , Análisis Multivariante , Dermopatía Fibrosante Nefrogénica/inducido químicamente , Dermopatía Fibrosante Nefrogénica/patología , Fenotipo , Factores de Riesgo
19.
Blood Adv ; 7(17): 5156-5171, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37417950

RESUMEN

Iron deficiency is a potent stimulator of fibroblast growth factor 23 (FGF23), a hormonal regulator of phosphate and vitamin D metabolism, that is classically thought to be produced by bone-embedded osteocytes. Here, we show that iron-deficient transmembrane serine protease 6 knockout (Tmprss6-/-) mice exhibit elevated circulating FGF23 and Fgf23 messenger RNA (mRNA) upregulation in the bone marrow (BM) but not the cortical bone. To clarify sites of Fgf23 promoter activity in Tmprss6-/- mice, we introduced a heterozygous enhanced green fluorescent protein (eGFP) reporter allele at the endogenous Fgf23 locus. Heterozygous Fgf23 disruption did not alter the severity of systemic iron deficiency or anemia in the Tmprss6-/- mice. Tmprss6-/-Fgf23+/eGFP mice showed green fluorescence in the vascular regions of BM sections and showed a subset of BM endothelial cells that were GFPbright by flow cytometry. Mining of transcriptomic data sets from mice with normal iron balance revealed higher Fgf23 mRNA in BM sinusoidal endothelial cells (BM-SECs) than that in other BM endothelial cell populations. Anti-GFP immunohistochemistry of fixed BM sections from Tmprss6-/-Fgf23+/eGFP mice revealed GFP expression in BM-SECs, which was more intense than in nonanemic controls. In addition, in mice with intact Tmprss6 alleles, Fgf23-eGFP reporter expression increased in BM-SECs following large-volume phlebotomy and also following erythropoietin treatment both ex vivo and in vivo. Collectively, our results identified BM-SECs as a novel site for Fgf23 upregulation in both acute and chronic anemia. Given the elevated serum erythropoietin in both anemic models, our findings raise the possibility that erythropoietin may act directly on BM-SECs to promote FGF23 production during anemia.


Asunto(s)
Anemia Ferropénica , Eritropoyetina , Animales , Ratones , Anemia Ferropénica/genética , Anemia Ferropénica/metabolismo , Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Eritropoyetina/genética , Eritropoyetina/metabolismo , Hierro , ARN Mensajero/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA