Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Cell Death Dis ; 10(6): 421, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142733

RESUMEN

Vacuole membrane protein (VMP1) is a putative autophagy protein, which together with Beclin-1 acts as a molecular switch in activating autophagy. In the present study the role of VMP1 was analysed in CD34+ cells of cord blood (CB) and primary acute myeloid leukemia (AML) cells and cell lines. An increased expression of VMP1 was observed in a subset of AML patients. Functional studies in normal CB CD34+ cells indicated that inhibiting VMP1 expression reduced autophagic-flux, coinciding with reduced expansion of hematopoietic stem and progenitor cells (HSPC), delayed differentiation, increased apoptosis and impaired in vivo engraftment. Comparable results were observed in leukemic cell lines and primary AML CD34+ cells. Ultrastructural analysis indicated that leukemic cells overexpressing VMP1 displayed a reduced number of mitochondrial structures, while the number of lysosomal degradation structures was increased. The overexpression of VMP1 did not affect cell proliferation and differentiation, but increased autophagic-flux and improved mitochondrial quality, which coincided with an increased threshold for venetoclax-induced loss of mitochondrial outer membrane permeabilization (MOMP) and apoptosis. In conclusion, our data indicate that in leukemic cells high VMP1 is involved with mitochondrial quality control.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteínas de la Membrana/metabolismo , Sulfonamidas/farmacología , Animales , Antígenos CD34/metabolismo , Puntos de Control del Ciclo Celular , Femenino , Sangre Fetal/citología , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Recambio Mitocondrial/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Sequestosoma-1/metabolismo
3.
Med Res Rev ; 39(2): 517-560, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30302772

RESUMEN

Autophagy is a crucial recycling process that is increasingly being recognized as an important factor in cancer initiation, cancer (stem) cell maintenance as well as the development of resistance to cancer therapy in both solid and hematological malignancies. Furthermore, it is being recognized that autophagy also plays a crucial and sometimes opposing role in the complex cancer microenvironment. For instance, autophagy in stromal cells such as fibroblasts contributes to tumorigenesis by generating and supplying nutrients to cancerous cells. Reversely, autophagy in immune cells appears to contribute to tumor-localized immune responses and among others regulates antigen presentation to and by immune cells. Autophagy also directly regulates T and natural killer cell activity and is required for mounting T-cell memory responses. Thus, within the tumor microenvironment autophagy has a multifaceted role that, depending on the context, may help drive tumorigenesis or may help to support anticancer immune responses. This multifaceted role should be taken into account when designing autophagy-based cancer therapeutics. In this review, we provide an overview of the diverse facets of autophagy in cancer cells and nonmalignant cells in the cancer microenvironment. Second, we will attempt to integrate and provide a unified view of how these various aspects can be therapeutically exploited for cancer therapy.


Asunto(s)
Autofagia , Neoplasias/patología , Microambiente Tumoral , Animales , Beclina-1/metabolismo , Neoplasias de la Mama/patología , Carcinogénesis , Transformación Celular Neoplásica , Femenino , Fibroblastos/metabolismo , Proteína HMGB1/metabolismo , Humanos , Hipoxia , Ratones , Mutación , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Células del Estroma/metabolismo
4.
Cell Death Dis ; 8(7): e2927, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28703806

RESUMEN

Here we have explored whether inhibition of autophagy can be used as a treatment strategy for acute myeloid leukemia (AML). Steady-state autophagy was measured in leukemic cell lines and primary human CD34+ AML cells with a large variability in basal autophagy between AMLs observed. The autophagy flux was higher in AMLs classified as poor risk, which are frequently associated with TP53 mutations (TP53mut), compared with favorable- and intermediate-risk AMLs. In addition, the higher flux was associated with a higher expression level of several autophagy genes, but was not affected by alterations in p53 expression by knocking down p53 or overexpression of wild-type p53 or p53R273H. AML CD34+ cells were more sensitive to the autophagy inhibitor hydroxychloroquine (HCQ) than normal bone marrow CD34+ cells. Similar, inhibition of autophagy by knockdown of ATG5 or ATG7 triggered apoptosis, which coincided with increased expression of p53. In contrast to wild-type p53 AML (TP53wt), HCQ treatment did not trigger a BAX and PUMA-dependent apoptotic response in AMLs harboring TP53mut. To further characterize autophagy in the leukemic stem cell-enriched cell fraction AML CD34+ cells were separated into ROSlow and ROShigh subfractions. The immature AML CD34+-enriched ROSlow cells maintained higher basal autophagy and showed reduced survival upon HCQ treatment compared with ROShigh cells. Finally, knockdown of ATG5 inhibits in vivo maintenance of AML CD34+ cells in NSG mice. These results indicate that targeting autophagy might provide new therapeutic options for treatment of AML since it affects the immature AML subfraction.


Asunto(s)
Autofagia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antígenos CD34/metabolismo , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteína 7 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Hidroxicloroquina/farmacología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/metabolismo
5.
Nucleic Acids Res ; 45(W1): W478-W483, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28472397

RESUMEN

Researchers usually query the large biomedical literature in PubMed via keywords, logical operators and filters, none of which is very intuitive. Question answering systems are an alternative to keyword searches. They allow questions in natural language as input and results reflect the given type of question, such as short answers and summaries. Few of those systems are available online but they experience drawbacks in terms of long response times and they support a limited amount of question and result types. Additionally, user interfaces are usually restricted to only displaying the retrieved information. For our Olelo web application, we combined biomedical literature and terminologies in a fast in-memory database to enable real-time responses to researchers' queries. Further, we extended the built-in natural language processing features of the database with question answering and summarization procedures. Combined with a new explorative approach of document filtering and a clean user interface, Olelo enables a fast and intelligent search through the ever-growing biomedical literature. Olelo is available at http://www.hpi.de/plattner/olelo.


Asunto(s)
Publicaciones , Programas Informáticos , Bases de Datos Factuales , Diabetes Mellitus/sangre , Brotes de Enfermedades , Internet , Lisofosfolípidos/sangre , PubMed , Esfingosina/análogos & derivados , Esfingosina/sangre , Interfaz Usuario-Computador , Infección por el Virus Zika/epidemiología
6.
Front Microbiol ; 7: 1633, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27818647

RESUMEN

Weak organic acids like sorbic and acetic acid are widely used to prevent growth of spoilage organisms such as Bacilli. To identify genes involved in weak acid stress tolerance we screened a transposon mutant library of Bacillus subtilis for sorbic acid sensitivity. Mutants of the rodZ (ymfM) gene were found to be hypersensitive to the lipophilic weak organic acid. RodZ is involved in determining the cell's rod-shape and believed to interact with the bacterial actin-like MreB cytoskeleton. Since rodZ lies upstream in the genome of the essential gene pgsA (phosphatidylglycerol phosphate synthase) we hypothesized that expression of the latter might also be affected in rodZ mutants and hence contribute to the phenotype observed. We show that both genes are co-transcribed and that both the rodZ::mini-Tn10 mutant and a conditional pgsA mutant, under conditions of minimal pgsA expression, were sensitive to sorbic and acetic acid. Both strains displayed a severely altered membrane composition. Compared to the wild-type strain, phosphatidylglycerol and cardiolipin levels were lowered and the average acyl chain length was elongated. Induction of rodZ expression from a plasmid in our transposon mutant led to no recovery of weak acid susceptibility comparable to wild-type levels. However, pgsA overexpression in the same mutant partly restored sorbic acid susceptibility and fully restored acetic acid sensitivity. A construct containing both rodZ and pgsA as on the genome led to some restored growth as well. We propose that RodZ and PgsA play intertwined roles in membrane homeostasis and tolerance to weak organic acid stress.

7.
Exp Hematol ; 44(12): 1188-1196.e6, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27616637

RESUMEN

ASXL1 is frequently mutated in myelodysplastic syndrome and other hematological malignancies. It has been reported that a loss of ASXL1 leads to a reduction of H3K27me3 via the polycomb repressive complex 2 (PRC2). To determine the role of ASXL1 loss in normal hematopoietic stem and progenitor cells, cord blood CD34+ cells were transduced with independent small hairpin interfering RNA lentiviral vectors against ASXL1 and cultured under myeloid and erythroid permissive conditions. Knockdown of ASXL1 led to a significant reduction in stem-cell frequency and a reduced cell expansion along the myeloid lineage. Cell expansion along the erythroid lineage was also reduced significantly and was accompanied by an increase in apoptosis of erythroid progenitor cells throughout differentiation and by an accumulation of cells in the G0/G1 phase. Bone marrow stromal cells supported the growth of immature erythroid cells, but did not alter the adverse phenotype of ASXL1 knockdown. Chromatin immunoprecipitation revealed no loss of H3K27me3 in myeloid progenitor cells, but demonstrated a loss of H3K27me3 on the HOXA and the p21 locus in erythroid progenitors. We conclude that ASXL1 is essential for erythroid development and differentiation and that the aberrant differentiation is, at least in part, facilitated via PRC2.


Asunto(s)
Apoptosis/genética , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Proteínas Represoras/deficiencia , Antígenos CD34/metabolismo , Biomarcadores , Comunicación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Precursoras Eritroides/metabolismo , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/citología , Histonas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Proteínas Represoras/genética
8.
Stem Cells ; 34(6): 1651-63, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26930546

RESUMEN

Autophagy is a highly regulated catabolic process that involves sequestration and lysosomal degradation of cytosolic components such as damaged organelles and misfolded proteins. While autophagy can be considered to be a general cellular housekeeping process, it has become clear that it may also play cell type-dependent functional roles. In this study, we analyzed the functional importance of autophagy in human hematopoietic stem/progenitor cells (HSPCs), and how this is regulated during differentiation. Western blot-based analysis of LC3-II and p62 levels, as well as flow cytometry-based autophagic vesicle quantification, demonstrated that umbilical cord blood-derived CD34(+) /CD38(-) immature hematopoietic progenitors show a higher autophagic flux than CD34(+) /CD38(+) progenitors and more differentiated myeloid and erythroid cells. This high autophagic flux was critical for maintaining stem and progenitor function since knockdown of autophagy genes ATG5 or ATG7 resulted in reduced HSPC frequencies in vitro as well as in vivo. The reduction in HSPCs was not due to impaired differentiation, but at least in part due to reduced cell cycle progression and increased apoptosis. This is accompanied by increased expression of p53, proapoptotic genes BAX and PUMA, and the cell cycle inhibitor p21, as well as increased levels of cleaved caspase-3 and reactive oxygen species. Taken together, our data demonstrate that autophagy is an important regulatory mechanism for human HSCs and their progeny, reducing cellular stress and promoting survival. Stem Cells 2016;34:1651-1663.


Asunto(s)
Antígenos CD34/metabolismo , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteína 7 Relacionada con la Autofagia/metabolismo , Autofagia , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Apoptosis , Recuento de Células , Ciclo Celular , Diferenciación Celular , Sangre Fetal/citología , Técnicas de Silenciamiento del Gen , Humanos , Espacio Intracelular/metabolismo , Ratones , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Especies Reactivas de Oxígeno/metabolismo
9.
Exp Hematol ; 43(3): 215-222.e2, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25462021

RESUMEN

To investigate whether the type of programmed cell death of myelodysplastic erythroid cells depends on their cellular context, we performed studies on cells from patients with low-risk myelodysplastic syndromes. We compared erythroid cells (and their precursor cells) from the mononuclear cell fraction with those from the hematon fraction, which are compacted complexes of hematopoietic cells surrounded by their own micro-environment. In directly fixed materials, erythroblasts exhibited signs of autophagy with limited apoptosis (<3%) based on ultrastructural characteristics and immunogold labeling for activated caspase-3. After 24 h in culture, myelodysplastic erythroblasts exhibited a significant increase in apoptosis (22 ± 7% vs. 3 ± 2%, p = 0.001). In contrast, the myelodysplastic erythroblasts from the hematon fraction did not exhibit an increased tendency toward apoptosis after culture (7 ± 3.3% vs. 1.8 ± 2.3%), which was in line with results for normal bone marrow cells. The same dependency on the micro-environment was noted for immature erythroid progenitor cells. Myelodysplastic hematons exhibited distinct numbers of erythroid burst-forming units in association with an extensive network of stromal cells, whereas small numbers of erythroid burst-forming units were generated from the myelodysplastic mononuclear cells compared with normal mononuclear cells (10.2 ± 9 vs. 162 ± 125, p < 0.001). Co-culture of erythroid myelodysplastic cells in the presence of growth factors (vascular endothelial growth factor, leukemia inhibitory factor) or on the MS-5 stromal layer did not restore the expansion of erythroid precursor cells. These data indicate that surviving myelodysplastic erythroid progenitors become more vulnerable to programmed cell death when they are detached from their own micro-environment.


Asunto(s)
Células Precursoras Eritroides/fisiología , Síndromes Mielodisplásicos/fisiopatología , Microambiente Tumoral , Anciano , Anciano de 80 o más Años , Apoptosis , Supervivencia Celular , Células Cultivadas , Células Precursoras Eritroides/patología , Femenino , Citometría de Flujo , Humanos , Leucocitos Mononucleares/patología , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Riesgo
10.
J Biol Chem ; 287(24): 20144-53, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22493507

RESUMEN

Peroxisomes play a major role in human cellular lipid metabolism, including fatty acid ß-oxidation. The most frequent peroxisomal disorder is X-linked adrenoleukodystrophy, which is caused by mutations in ABCD1. The biochemical hallmark of X-linked adrenoleukodystrophy is the accumulation of very long chain fatty acids (VLCFAs) due to impaired peroxisomal ß-oxidation. Although this suggests a role of ABCD1 in VLCFA import into peroxisomes, no direct experimental evidence is available to substantiate this. To unravel the mechanism of peroxisomal VLCFA transport, we use Saccharomyces cerevisiae as a model organism. Here we provide evidence that in this organism very long chain acyl-CoA esters are hydrolyzed by the Pxa1p-Pxa2p complex prior to the actual transport of their fatty acid moiety into the peroxisomes with the CoA presumably being released into the cytoplasm. The Pxa1p-Pxa2p complex functionally interacts with the acyl-CoA synthetases Faa2p and/or Fat1p on the inner surface of the peroxisomal membrane for subsequent re-esterification of the VLCFAs. Importantly, the Pxa1p-Pxa2p complex shares this molecular mechanism with HsABCD1 and HsABCD2.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de Transporte de Ácidos Grasos/metabolismo , Ácidos Grasos/metabolismo , Peroxisomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Subfamilia D de Transportadores de Casetes de Unión al ATP , Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/genética , Adrenoleucodistrofia/genética , Adrenoleucodistrofia/metabolismo , Transporte Biológico Activo/fisiología , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Proteínas de Transporte de Ácidos Grasos/genética , Ácidos Grasos/genética , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Oxidación-Reducción , Peroxisomas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA