Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo de estudio
País/Región como asunto
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Mycologia ; 106(2): 216-23, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24782491

RESUMEN

We document here for the first time ultrastructural details of the cellular interaction of Asteridiella callista and its host Stachytarpheta mutabilis var. violacea from Costa Rica. A. callista attaches to the host with appressoria, invades the epidermal cell wall and forms an apoplastic complex cisternal net, presumably for nutrient uptake from its host. This unique structure, called an interaction apparatus (Ia), consists of cisternae surrounded by a membrane continuous with the fungal cytoplasmic membrane. Subsequently the apoplastic trunk of the Ia extends into the host epidermal cell wall and contacts the host cytoplasmic membrane. Electron-opaque material, probably of fungal origin appears at the host cytoplasmic membrane. Finally these electron-opaque deposits are encased by host material. Functional and systematical aspects of this interaction scenario are discussed.


Asunto(s)
Ascomicetos/fisiología , Enfermedades de las Plantas/microbiología , Verbenaceae/microbiología , Ascomicetos/crecimiento & desarrollo , Ascomicetos/ultraestructura , Costa Rica , Hifa/crecimiento & desarrollo , Hifa/fisiología , Hifa/ultraestructura , Verbenaceae/fisiología
2.
Cancers (Basel) ; 14(10)2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35625953

RESUMEN

Despite advances in available treatments, multiple myeloma (MM) remains an incurable disease and represents a challenge in oncohematology. New insights into epigenetic factors contributing to MM development and progression have improved the knowledge surrounding its molecular basis. Beyond classical epigenetic factors, including methylation and acetylation, recent genome analyses have unveiled the importance of non-coding RNAs in MM pathogenesis. Non-coding RNAs have become of interest, as their dysregulation opens the door to new therapeutic approaches. The discovery, in the past years, of molecular techniques, such as CRISPR-Cas, has led to innovative therapies with potential benefits to achieve a better outcome for MM patients. This review summarizes the current knowledge on epigenetics and non-coding RNAs in MM pathogenesis.

3.
Front Immunol ; 12: 723836, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630398

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory disorder. HLH can be considered as a threshold disease depending on the trigger and the residual NK-cell cytotoxicity. In this study, we analyzed the molecular and functional impact of a novel monoallelic mutation found in a patient with two episodes of HLH. A 9-month-old child was diagnosed at 2 months of age with cutaneous Langerhans cell histiocytosis (LCH). After successful treatment, the patient developed an HLH episode. At 16 month of age, the patient went through an HSCT losing the engraftment 5 months later concomitant with an HLH relapse. The genetic study revealed a monoallelic mutation in the STXBP2 gene (.pArg190Cys). We transfected COS7 cells to analyze the STXBP2-R190C expression and to test the interaction with STX11. We used the RBL-2H3 cell line expressing STXBP2-WT-EGFP or R190C-EGFP for degranulation assays. Mutation STXBP2-R190C did not affect protein expression or interaction with syntaxin-11. However, we have demonstrated that STXBP2-R190C mutation diminishes degranulation in the RBL-2H3 cell line compared with the RBL-2H3 cell line transfected with STXBP2-WT or nontransfected. These results suggest that STXBP2-R190C mutation acts as a modifier of the degranulation process producing a decrease in degranulation. Therefore, under homeostatic conditions, the presence of one copy of STXBP2-R190 could generate sufficient degranulation capacity. However, it is likely that early in life when adaptive immune system functions are not sufficiently developed, an infection may not be resolved with this genetic background, leading to a hyperinflammation syndrome and eventually develop HLH. This analysis highlights the need for functional testing of new mutations to validate their role in genetic susceptibility and to establish the best possible treatment for these patients.


Asunto(s)
Histiocitosis de Células de Langerhans/diagnóstico , Histiocitosis de Células de Langerhans/genética , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/genética , Proteínas Munc18/genética , Citotoxicidad Inmunológica , Predisposición Genética a la Enfermedad , Histiocitosis de Células de Langerhans/complicaciones , Humanos , Lactante , Linfohistiocitosis Hemofagocítica/complicaciones , Masculino , Mutación
4.
Int J Hematol ; 111(3): 440-450, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31865540

RESUMEN

The presence of mutations in PRF1, UNC13D, STX11 and STXBP2 genes in homozygosis or compound heterozygosis results in immune deregulation. Most such cases lead to clinical manifestations of haemophagocytic lymphohistiocytosis (HLH). In the present study, we analyzed degranulation and cytotoxicity in a pediatric patient with a late presentation of HLH associated with Epstein-Barr virus infection. Remarkably, the results of the degranulation assay showed reduction of CD107a median fluorescence intensity (MFI) and absent cytotoxicity. Genetic analysis identified compound heterozygous mutations in STXBP2 gene: a previously reported splicing defect in exon 15 (c.1247-1G>C, p.V417LfsX126) and a novel missense mutation in exon 9 (c.728T>G, p.L243R). Transfection experiments of STXBP2-L243R or STXBP2-WT constructs showed an undetectable protein expression of the STXBP2-L243R mutation. The residue L243 is highly preserved evolutionarily; moreover, computational analysis of its structure revealed its participation in the rich network of interactions that stabilizes domains 2 and 3 of the protein. Altogether, we demonstrated by molecular and in silico analysis that the new L243R mutation in STXBP2 plays a pathogenic role that, together with the p.Val417Leufsc mutation, shows the synergistic negative effect of these two mutations on STXBP2 function, leading to a decrease of degranulatory activity in vivo.


Asunto(s)
Degranulación de la Célula , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/patología , Proteínas Munc18/genética , Mutación , Animales , Células COS , Preescolar , Chlorocebus aethiops , Infecciones por Virus de Epstein-Barr/complicaciones , Humanos , Linfohistiocitosis Hemofagocítica/complicaciones , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA