Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 12(8)2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32764479

RESUMEN

Cell fate decisions regulating survival and death are essential for maintaining tissue homeostasis; dysregulation thereof can lead to tumor development. In some cases, survival and death are triggered by the same receptor, e.g., tumor necrosis factor (TNF)-receptor 1 (TNFR1). We identified a prominent role for the cold shock Y-box binding protein-1 (YB-1) in the TNF-induced activation and nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65. In the absence of YB-1, the expression of TNF receptor-associated factor 2 (TRAF2), a central component of the TNF receptor signaling complex required for NF-κB activation, is significantly reduced. Therefore, we hypothesized that the loss of YB-1 results in a destabilization of TRAF2. Consistent with this hypothesis, we observed that YB-1-deficient cells were more prone to TNF-induced apoptotic cell death. We observed enhanced effector caspase-3 activation and could successfully rescue the cells using the pan-caspase inhibitor zVAD-fmk, but not necrostatin-1. Taken together, our results indicate that YB-1 plays a central role in promoting cell survival through NF-κB activation and identifies a novel mechanism by which enhanced YB-1 expression may contribute to tumor development.

2.
BMC Genomics ; 20(Suppl 3): 293, 2019 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-31815628

RESUMEN

BACKGROUND: Structural homology modeling supported by bioinformatics analysis plays a key role in uncovering new molecular interactions within gene regulatory networks. Here, we have applied this powerful approach to analyze the molecular interactions orchestrating death receptor signaling networks. In particular, we focused on the molecular mechanisms of CD95-mediated NF-κB activation and the role of c-FLIP/NEMO interaction in the induction of this pathway. RESULTS: To this end, we have created the homology model of the c-FLIP/NEMO complex using the reported structure of the v-FLIP/NEMO complex, and rationally designed peptides targeting this complex. The designed peptides were based on the NEMO structure. Strikingly, the experimental in vitro validation demonstrated that the best inhibitory effects on CD95-mediated NF-κB activation are exhibited by the NEMO-derived peptides with the substitution D242Y of NEMO. Furthermore, we have assumed that the c-FLIP/NEMO complex is recruited to the DED filaments formed upon CD95 activation and validated this assumption in silico. Further insight into the function of c-FLIP/NEMO complex was provided by the analysis of evolutionary conservation of interacting regions which demonstrated that this interaction is common in distinct mammalian species. CONCLUSIONS: Taken together, using a combination of bioinformatics and experimental approaches we obtained new insights into CD95-mediated NF-κB activation, providing manifold possibilities for targeting the death receptor network.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Quinasa I-kappa B/metabolismo , Sondas Moleculares , FN-kappa B/metabolismo , Receptor fas/metabolismo , Secuencia de Aminoácidos , Biología Computacional , Humanos , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Alineación de Secuencia , Transducción de Señal
3.
PLoS Comput Biol ; 14(9): e1006368, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30256782

RESUMEN

CD95/Fas/APO-1 is a member of the death receptor family that triggers apoptotic and anti-apoptotic responses in particular, NF-κB. These responses are characterized by a strong heterogeneity within a population of cells. To determine how the cell decides between life and death we developed a computational model supported by imaging flow cytometry analysis of CD95 signaling. Here we show that CD95 stimulation leads to the induction of caspase and NF-κB pathways simultaneously in one cell. The related life/death decision strictly depends on cell-to-cell variability in the formation of the death-inducing complex (DISC) on one side (extrinsic noise) vs. stochastic gene expression of the NF-κB pathway on the other side (intrinsic noise). Moreover, our analysis has uncovered that the stochasticity in apoptosis and NF-kB pathways leads not only to survival or death of a cell, but also causes a third type of response to CD95 stimulation that we termed ambivalent response. Cells in the ambivalent state can undergo cell death or survive which was subsequently validated by experiments. Taken together, we have uncovered how these two competing pathways control the fate of a cell, which in turn plays an important role for development of anti-cancer therapies.


Asunto(s)
Análisis de la Célula Individual/métodos , Receptor fas/fisiología , Apoptosis , Caspasa 3/metabolismo , Caspasas/metabolismo , Linaje de la Célula , Simulación por Computador , Citometría de Flujo , Células HeLa , Humanos , Modelos Teóricos , FN-kappa B/metabolismo , Transducción de Señal
4.
Int J Biochem Cell Biol ; 102: 101-108, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30025878

RESUMEN

The role of caspase-2 in cell death regulation remains largely unknown. In this study we have analyzed the involvement of caspase-2 in RIPK1-regulated necrosis (necroptosis) in human ovarian carcinoma cells. We show that these cells undergo necroptosis upon treatment with the DNA damaging drug cisplatin in combination with the pan-caspase inhibitor zVAD-fmk. Downregulation of caspase-2 leads to an increase of necroptosis in CAOV-4 cells. Interestingly, an association of caspase-2 to the necrosome complex was not detected. Importantly, downregulation of caspase-2 with shRNA or CRISPR/Cas9 system led to an enhanced phosphorylation of RIPK1 and MLKL. Taken together, our data strongly indicate that caspase-2 negatively regulates necroptotic cell death, which might play an important role in further therapeutic applications.


Asunto(s)
Caspasa 2/metabolismo , Línea Celular , Línea Celular Tumoral , Humanos , Terapia Molecular Dirigida , Necrosis/enzimología , Transporte de Proteínas
5.
PLoS One ; 13(5): e0197208, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29768460

RESUMEN

Imaging flow cytometry is a powerful experimental technique combining the strength of microscopy and flow cytometry to enable high-throughput characterization of cell populations on a detailed microscopic scale. This approach has an increasing importance for distinguishing between different cellular phenotypes such as proliferation, cell division and cell death. In the course of undergoing these different pathways, each cell is characterized by a high amount of properties. This makes it hard to filter the most relevant information for cell state discrimination. The traditional methods for cell state discrimination rely on dye based two-dimensional gating strategies ignoring information that is hidden in the high-dimensional property space. In order to make use of the information ignored by the traditional methods, we present a simple and efficient approach to distinguish biological states within a cell population based on machine learning techniques. We demonstrate the advantages and drawbacks of filter techniques combined with different classification schemes. These techniques are illustrated with two case studies of apoptosis detection in HeLa cells. Thereby we highlight (i) the aptitude of imaging flow cytometry regarding automated, label-free cell state discrimination and (ii) pitfalls that are frequently encountered. Additionally a MATLAB script is provided, which gives further insight regarding the computational work presented in this study.


Asunto(s)
Apoptosis , Citometría de Flujo/métodos , Animales , Citometría de Flujo/instrumentación , Células HeLa , Humanos
6.
Bio Protoc ; 7(1): e2081, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34458412

RESUMEN

Apoptosis or programmed cell death is important for multicellular organisms to keep cell homeostasis and for the clearance of mutated or infected cells. Apoptosis can be induced by intrinsic or extrinsic stimuli. The first event in extrinsic apoptosis is the formation of the Death-Inducing Signalling Complex (DISC), where the initiator caspases-8 and -10 are fully activated by several proteolytic cleavage steps and induce the caspase cascade leading to apoptotic cell death. Analysing the processing of procaspases-8 and -10 by Western blot is a commonly used method to study the induction of apoptosis by death receptor stimulation. To analyse procaspase-8 and -10 cleavage, cells are stimulated with a death ligand for different time intervals, lysed and subjected to Western blot analysis using anti-caspase-8 and anti-caspase-10 antibodies. This allows monitoring the caspase cleavage products and thereby induction of apoptosis.

7.
Sci Rep ; 6: 39796, 2016 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-28004776

RESUMEN

The ubiquitin-modifying enzyme A20, an important negative feedback regulator of NF-κB, impairs the expansion of tumor-specific CD8+ T cells but augments the proliferation of autoimmune CD4+ T cells. To study the T cell-specific function of A20 in bacterial infection, we infected T cell-specific A20 knockout (CD4-Cre A20fl/fl) and control mice with Listeria monocytogenes. A20-deficient pathogen-specific CD8+ T cells expanded stronger resulting in improved pathogen control at day 7 p.i. Imaging flow cytometry revealed that A20-deficient Listeria-specific CD8+ T cells underwent increased apoptosis and necroptosis resulting in reduced numbers of memory CD8+ T cells. In contrast, the primary CD4+ T cell response was A20-independent. Upon secondary infection, the increase and function of pathogen-specific CD8+ T cells, as well as pathogen control were significantly impaired in CD4-Cre A20fl/fl mice. In vitro, apoptosis and necroptosis of Listeria-specific A20-deficient CD8+ T cells were strongly induced as demonstrated by increased caspase-3/7 activity, RIPK1/RIPK3 complex formation and more morphologically apoptotic and necroptotic CD8+ T cells. In vitro, A20 limited CD95L and TNF-induced caspase3/7 activation. In conclusion, T cell-specific A20 limited the expansion but reduced apoptosis and necroptosis of Listeria-specific CD8+ T cells, resulting in an impaired pathogen control in primary but improved clearance in secondary infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Listeria/inmunología , Listeriosis/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Memoria Inmunológica , Listeriosis/genética , Listeriosis/patología , Ratones , Ratones Transgénicos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...