Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 134(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34806752

RESUMEN

Extrinsic apoptosis relies on TNF-family receptor activation by immune cells or receptor-activating drugs. Here, we monitored cell cycle progression at a resolution of minutes to relate apoptosis kinetics and cell-to-cell heterogeneities in death decisions to cell cycle phases. Interestingly, we found that cells in S phase delay TRAIL receptor-induced death in favour of mitosis, thereby passing on an apoptosis-primed state to their offspring. This translates into two distinct fates, apoptosis execution post mitosis or cell survival from inefficient apoptosis. Transmitotic resistance is linked to Mcl-1 upregulation and its increased accumulation at mitochondria from mid-S phase onwards, which allows cells to pass through mitosis with activated caspase-8, and with cells escaping apoptosis after mitosis sustaining sublethal DNA damage. Antagonizing Mcl-1 suppresses cell cycle-dependent delays in apoptosis, prevents apoptosis-resistant progression through mitosis and averts unwanted survival after apoptosis induction. Cell cycle progression therefore modulates signal transduction during extrinsic apoptosis, with Mcl-1 governing decision making between death, proliferation and survival. Cell cycle progression thus is a crucial process from which cell-to-cell heterogeneities in fates and treatment outcomes emerge in isogenic cell populations during extrinsic apoptosis. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Apoptosis , Transducción de Señal , Ciclo Celular , Línea Celular Tumoral , Humanos , Mitosis , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF
2.
J Biol Chem ; 289(23): 16576-87, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24764293

RESUMEN

The cytokine TNF-related apoptosis-inducing ligand (TRAIL) and its cell membrane receptors constitute an elaborate signaling system fulfilling important functions in immune regulation and tumor surveillance. Activation of the death receptors TRAILR1 and TRAILR2 can lead to apoptosis, whereas TRAILR3 and TRAILR4 are generally referred to as decoy receptors, which have been shown to inhibit TRAIL-induced apoptosis. The underlying molecular mechanisms, however, remain unclear. Alike other members of the TNF receptor superfamily, TRAIL receptors contain a pre-ligand binding assembly domain (PLAD) mediating receptor oligomerization. Still, the stoichiometry of TRAIL receptor oligomers as well as the issue of whether the PLAD mediates only homotypic or also heterotypic interactions remained inconclusive until now. Performing acceptor-photobleaching FRET studies with receptors 1, 2, and 4, we demonstrate interactions in all possible combinations. Formation of dimers was shown by chemical cross-linking experiments for interactions of TRAILR2 and heterophilic interactions between the two death receptors or between either of the death receptors and TRAILR4. Implications of the demonstrated receptor-receptor interactions on signaling were investigated in suitable cellular models. Both apoptosis induction and activation of the transcription factor NFκB were significantly reduced in the presence of TRAILR4. Our experimental data combined with mathematical modeling show that the inhibitory capacity of TRAILR4 is attributable to signaling-independent mechanisms, strongly suggesting a reduction of signaling competent death receptors through formation heteromeric receptor complexes. In summary, we propose a model of TRAIL receptor interference driven by PLAD-mediated formation of receptor heterodimers on the cell membrane.


Asunto(s)
Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Transducción de Señal , Apoptosis , Dimerización , Células HeLa , Humanos
3.
BMC Bioinformatics ; 12: 125, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21527025

RESUMEN

BACKGROUND: Most of the modeling performed in the area of systems biology aims at achieving a quantitative description of the intracellular pathways within a "typical cell". However, in many biologically important situations even clonal cell populations can show a heterogeneous response. These situations require study of cell-to-cell variability and the development of models for heterogeneous cell populations. RESULTS: In this paper we consider cell populations in which the dynamics of every single cell is captured by a parameter dependent differential equation. Differences among cells are modeled by differences in parameters which are subject to a probability density. A novel Bayesian approach is presented to infer this probability density from population snapshot data, such as flow cytometric analysis, which do not provide single cell time series data. The presented approach can deal with sparse and noisy measurement data. Furthermore, it is appealing from an application point of view as in contrast to other methods the uncertainty of the resulting parameter distribution can directly be assessed. CONCLUSIONS: The proposed method is evaluated using artificial experimental data from a model of the tumor necrosis factor signaling network. We demonstrate that the methods are computationally efficient and yield good estimation result even for sparse data sets.


Asunto(s)
Teorema de Bayes , Técnicas Citológicas , Modelos Biológicos , Análisis de Regresión , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
4.
Biochim Biophys Acta ; 1798(6): 1081-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20188063

RESUMEN

The proinflammatory cytokine tumor necrosis factor (TNF) binds two distinct plasma membrane receptors, TNFR1 and TNFR2. We have produced different receptor mutants fused with enhanced green fluorescent protein to study their membrane dynamics by fluorescence correlation spectroscopy (FCS). TNFR1 mutants show diffusion constants of approximately 1.2 x10(-9)cm(2)/s and a broad distribution of diffusion times, which is hardly affected by ligand binding. However, cholesterol depletion enhances their diffusion, suggesting a constitutive affinity to cholesterol rich membrane microdomains. In contrast, TNFR2 and mutants thereof diffuse rather fast (D=3.1 x10(-9)cm(2)/s) with a marked reduction after 30 min of TNF treatment (D=0.9 x 10(-9)cm(2)/s). This reduction cannot be explained by the formation of higher ordered receptor clusters, since the fluorescence intensity of TNF treated receptors indicate the presence of a few receptor molecules per complex only. Together, these data point to a topological segregation of the two TNF receptors in different microcompartments of the plasma membrane independent of the cytoplasmic signaling domains of the receptors.


Asunto(s)
Microdominios de Membrana/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ligandos , Microdominios de Membrana/genética , Ratones , Ratones Noqueados , Mutación , Estructura Terciaria de Proteína , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Transducción de Señal/fisiología , Espectrometría de Fluorescencia , Factor de Necrosis Tumoral alfa/metabolismo
5.
PLoS Comput Biol ; 5(12): e1000595, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20011108

RESUMEN

Apoptosis is regulated by several signaling pathways which are extensively linked by crosstalks. Boolean or logical modeling has become a promising approach to capture the qualitative behavior of such complex networks. Here we built a large-scale literature-based Boolean model of the central intrinsic and extrinsic apoptosis pathways as well as pathways connected with them. The model responds to several external stimuli such as Fas ligand, TNF-alpha, UV-B irradiation, interleukin-1beta and insulin. Timescales and multi-value node logic were used and turned out to be indispensable to reproduce the behavior of the apoptotic network. The coherence of the model was experimentally validated. Thereby an UV-B dose-effect is shown for the first time in mouse hepatocytes. Analysis of the model revealed a tight regulation emerging from high connectivity and spanning crosstalks and a particular importance of feedback loops. An unexpected feedback from Smac release to RIP could further increase complex II formation. The introduced Boolean model provides a comprehensive and coherent description of the apoptosis network behavior. It gives new insights into the complex interplay of pro- and antiapoptotic factors and can be easily expanded to other signaling pathways.


Asunto(s)
Apoptosis , Animales , Apoptosis/efectos de la radiación , Biología Computacional , Humanos , Ratones , Modelos Biológicos , Transducción de Señal
6.
Nano Lett ; 9(12): 4240-5, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19772290

RESUMEN

Cellular apoptosis, the prototype of programmed cell death, can be induced by activation of so-called death receptors. Interestingly, soluble and membrane-bound members of death receptor ligands can differentially activate their receptors. Using the death receptor ligand tumor necrosis factor (TNF) presented on a surface in a nanoscaled pattern with spacings between 58 and 290 nm, we investigated its requirements for spatial arrangement and motility to efficiently activate TNF receptor (TNFR)1 and TNFR2 as well as its chimeras TNFR1-Fas and TNFR2-Fas. We show that the mere mechanical fixation of TNF is insufficient to efficiently activate TNFR2 that is responsive to only the membrane bound form of TNF but not its soluble form. Rather, an additional stabilization of TNFR2(-Fas) by cluster formation seems to be mandatory for efficient activation. In contrast, TNFR1(-Fas) is strongly activated by TNF spaced within up to 200 nm distances, whereas larger spacings of 290 nm fails completely. Furthermore, unlike for TNFR2(-Fas) no dose-response relationship to increasing distances of nanostructured ligands could be observed for TNFR1-(Fas), suggesting that compartmentalization of the cell membrane in confinement zones of approximately 200 nm regulates TNFR1 activation.


Asunto(s)
Apoptosis/fisiología , Fibroblastos/fisiología , Nanoestructuras/administración & dosificación , Nanoestructuras/química , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/farmacología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Fibroblastos/efectos de los fármacos , Ensayo de Materiales , Ratones , Propiedades de Superficie
7.
Cell Death Differ ; 27(11): 3037-3052, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32433558

RESUMEN

The influence of 3D microenvironments on apoptosis susceptibility remains poorly understood. Here, we studied the susceptibility of cancer cell spheroids, grown to the size of micrometastases, to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Interestingly, pronounced, spatially coordinated response heterogeneities manifest within spheroidal microenvironments: In spheroids grown from genetically identical cells, TRAIL-resistant subpopulations enclose, and protect TRAIL-hypersensitive cells, thereby increasing overall treatment resistance. TRAIL-resistant layers form at the interface of proliferating and quiescent cells and lack both TRAILR1 and TRAILR2 protein expression. In contrast, oxygen, and nutrient deprivation promote high amounts of TRAILR2 expression in TRAIL-hypersensitive cells in inner spheroid layers. COX-II inhibitor celecoxib further enhanced TRAILR2 expression in spheroids, likely resulting from increased ER stress, and thereby re-sensitized TRAIL-resistant cell layers to treatment. Our analyses explain how TRAIL response heterogeneities manifest within well-defined multicellular environments, and how spatial barriers of TRAIL resistance can be minimized and eliminated.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Esferoides Celulares/patología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Celecoxib/farmacología , Línea Celular Tumoral , Inhibidores de la Ciclooxigenasa 2/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
8.
J Cell Biol ; 166(3): 369-80, 2004 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-15289496

RESUMEN

Fas (APO-1/CD95) is the prototypic death receptor, and the molecular mechanisms of Fas-induced apoptosis are comparably well understood. Here, we show that Fas activates NFkappaB via a pathway involving RIP, FADD, and caspase-8. Remarkably, the enzymatic activity of the latter was dispensable for Fas-induced NFkappaB signaling pointing to a scaffolding-related function of caspase-8 in nonapoptotic Fas signaling. NFkappaB was activated by overexpressed FLIPL and FLIPS in a cell type-specific manner. However, in the context of Fas signaling both isoforms blocked FasL-induced NFkappaB activation. Moreover, down-regulation of both endogenous FLIP isoforms or of endogenous FLIPL alone was sufficient to enhance FasL-induced expression of the NFkappaB target gene IL8. As NFkappaB signaling is inhibited during apoptosis, FasL-induced NFkappaB activation was most prominent in cells that were protected by Bcl2 expression or caspase inhibitors and expressed no or minute amounts of FLIP. Thus, protection against Fas-induced apoptosis in a FLIP-independent manner converted a proapoptotic Fas signal into an inflammatory NFkappaB-related response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , FN-kappa B/metabolismo , Proteínas/metabolismo , Receptor fas/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Caspasa 8 , Proteína de Dominio de Muerte Asociada a Fas , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas I-kappa B/metabolismo , Inhibidor NF-kappaB alfa , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Regulación hacia Arriba
9.
MAbs ; 11(1): 166-177, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30252601

RESUMEN

Selective inhibition of tumor necrosis factor (TNF) signaling through the proinflammatory axis of TNF-receptor 1 (TNFR1) while leaving pro-survival and regeneration-promoting signals via TNFR2 unaffected is a promising strategy to circumvent limitations of complete inhibition of TNF action by the approved anti-TNF drugs. A previously developed humanized antagonistic TNFR1-specific antibody, ATROSAB, showed potent inhibition of TNFR1-mediated cellular responses. Because the parental mouse antibody H398 possesses even stronger inhibitory potential, we scrutinized the specific binding parameters of the two molecules and revealed a faster dissociation of ATROSAB compared to H398. Applying affinity maturation and re-engineering of humanized variable domains, we generated a monovalent Fab derivative (13.7) of ATROSAB that exhibited increased binding to TNFR1 and superior inhibition of TNF-mediated TNFR1 activation, while lacking any agonistic activity even in the presence of cross-linking antibodies. In order to improve its pharmacokinetic properties, several Fab13.7-derived molecules were generated, including a PEGylated Fab, a mouse serum albumin fusion protein, a half-IgG with a dimerization-deficient Fc, and a newly designed Fv-Fc format, employing the knobs-into-holes technology. Among these derivatives, the Fv13.7-Fc displayed the best combination of improved pharmacokinetic properties and antagonistic activity, thus representing a promising candidate for further clinical development.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Anticuerpos de Cadena Única/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos , Humanos , Ratones , Ingeniería de Proteínas/métodos
10.
PLoS One ; 13(6): e0198203, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29927992

RESUMEN

Dysregulation of the mitochondrial signaling pathway of apoptosis induction represents a major hurdle in tumor therapy. The objective of the presented work was to investigate the role of the intrinsic (mitochondrial) apoptotic pathway in the non-small lung cancer cell line NCI-H460 upon induction of apoptosis using the highly bioactive TRAIL derivative Db-scTRAIL. NCI-H460 cells were TRAIL sensitive but an only about 3 fold overexpression of Bcl-2 was sufficient to induce a highly TRAIL resistant phenotype, confirming that the mitochondrial pathway is crucial for TRAIL-induced apoptosis induction. TRAIL resistance was paralleled by a strong inhibition of caspase-8, -9 and -3 activities and blocked their full processing. Notably, especially the final cleavage steps of the initiator caspase-8 and the executioner caspase-3 were effectively blocked by Bcl-2 overexpression. Caspase-9 knockdown failed to protect NCI-H460 cells from TRAIL-induced cell death, suggesting a minor role of this initiator caspase in this apoptotic pathway. Rather, knockdown of the XIAP antagonist Smac resulted in enhanced caspase-3 degradation after stimulation of cells with TRAIL. Of note, downregulation of XIAP had only limited effects on TRAIL sensitivity of wild-type NCI-H460 cells, but resensitized Bcl-2 overexpressing cells for TRAIL-induced apoptosis. In particular, XIAP knockdown in combination with TRAIL allowed the final cleavage step of caspase-3 to generate the catalytically active p17 fragment, whose production was otherwise blocked in Bcl-2 overexpressing cells. Together, our data strongly suggest that XIAP-mediated inhibition of final caspase-3 processing is the last and major hurdle in TRAIL-induced apoptosis in NCI-H460 cells, which can be overcome by Smac in a Bcl-2 level dependent manner. Quantitative investigation of the XIAP/Smac interplay using a mathematical model approach corroborates our experimental data strengthening the suggested roles of XIAP and Smac as critical determinants for TRAIL sensitivity.


Asunto(s)
Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Caspasas/metabolismo , Neoplasias Pulmonares/metabolismo , Modelos Biológicos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Caspasas/genética , Células HCT116 , Células HeLa , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
11.
Cytokine Growth Factor Rev ; 15(5): 353-66, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15450251

RESUMEN

The inflammatory cytokine tumor necrosis factor (TNF), as well as most other ligand members of the TNF superfamily, exist both as classical soluble cytokines, but also in the form of type II transmembrane proteins. Both forms possess bioactivity, although some effects are distinct. In addition, an increasing body of evidence suggests that the membrane integrated ligands can receive signals, i.e. act as receptors which can transmit positive and negative feedback signals into the ligand bearing cell. Thus, reverse signaling enables a two-way communication in cell-to-cell signaling, and it is conceivable that this bi-directional signal exchange contributes to the plasticity of the ligand-receptor systems. Reverse signaling mainly has been observed in the immune system and within the TNF superfamily. Its function is only beginning to emerge warranting additional investigation, especially when it comes to therapeutic strategies involving cytokine modulation. This review provides an update of the literature about reverse signaling of transmembrane TNF family members and discusses its potential biological and clinical impact.


Asunto(s)
Antígenos CD/metabolismo , Sistema Inmunológico/fisiología , Proteínas de la Membrana/metabolismo , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adaptación Fisiológica/fisiología , Animales , Citocinas/metabolismo , Humanos , Ligandos , Receptores de Citocinas/metabolismo , Transducción de Señal/fisiología , Relación Estructura-Actividad
12.
Cytokine Growth Factor Rev ; 14(1): 53-66, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12485619

RESUMEN

Fas (Apo-1, CD95) and Fas-Ligand (FasL, CD95L) are typical members of the TNF receptor and TNF ligand family, respectively, with a pivotal role in the regulation of apoptotic processes, including activation-induced cell death, T-cell-induced cytotoxicity, immune privilege and tumor surveillance. Impairment of the FasL/Fas system has been implicated in liver failure, autoimmune diseases and immune deficiency. Thus, the FasL/Fas system was mainly appreciated with respect to its death-inducing capabilities. However, there is increasing evidence that activation of Fas can also result in non-apoptotic responses like cell proliferation or NF-kappaB activation. While the apoptotic features of the FasL/Fas system and the pathways involved are comparably well investigated, the pathways that are utilized by Fas to transduce proliferative and activating signals are poorly understood. This review is focused on the non-apoptotic functions of the FasL/Fas system. In particular, the similarities and differences of the molecular mechanisms of apoptotic and non-apoptotic Fas signaling are addressed.


Asunto(s)
Apoptosis/fisiología , Transducción de Señal/fisiología , Receptor fas/metabolismo , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Activación Enzimática , Proteína Ligando Fas , Humanos , Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Glicoproteínas de Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo
13.
Biosystems ; 90(3): 591-601, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17314003

RESUMEN

Signal transduction networks are complex, as are their mathematical models. Gaining a deeper understanding requires a system analysis. Important aspects are the number, location and stability of steady states. In particular, bistability has been recognised as an important feature to achieve molecular switching. This paper compares different model structures and analysis methods particularly useful for bistability analysis. The biological applications include proteolytic cascades as, for example, encountered in the apoptotic signalling pathway or in the blood clotting system. We compare three model structures containing zero-order, inhibitor and cooperative ultrasensitive reactions, all known to achieve bistability. The combination of phase plane and bifurcation analysis provides an illustrative and comprehensive understanding of how bistability can be achieved and indicates how robust this behaviour is. Experimentally, some so-called "inactive" components were shown to have a residual activity. This has been mostly ignored in mathematical models. Our analysis reveals that bistability is only mildly affected in the case of zero-order or inhibitor ultrasensitivity. However, the case where bistability is achieved by cooperative ultrasensitivity is severely affected by this perturbation.


Asunto(s)
Modelos Biológicos , Péptido Hidrolasas/metabolismo , Precursores Enzimáticos/metabolismo , Estabilidad de Enzimas , Matemática , Transducción de Señal , Biología de Sistemas
14.
Perit Dial Int ; 37(3): 250-258, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28096440

RESUMEN

♦ BACKGROUND: Peritoneal dialysis (PD) coincides with high concentrations of proinflammatory cytokines, such as tumor necrosis factor (TNF), in the peritoneal cavity. During treatment, chronic inflammatory processes lead to damage of the peritoneal membrane and a subsequent ultrafiltration failure. Human peritoneal mesothelial cells (HPMCs) play a central role as mediators and targets of PD-related inflammatory changes. Although TNF Receptor 1 (TNFR1) is expressed in high numbers on the cells, TNF-induced apoptosis is inhibited. Here, the underlying molecular mechanisms of TNFR1 signaling in HPMCs are investigated. ♦ METHODS: Human peritoneal mesothelial cells were isolated from the omentum of healthy donors and the dialysis solution of PD patients. Flow cytometry was applied to determine cell surface expression of TNFR1 on HPMCS from healthy donors in absence or presence of TNF or PD fluid (PDF) and were compared to TNFR1 expression on cells from PD patients. To investigate TNFR1-mediated signaling, HPMCs were treated with PDF or TNF, and expression patterns of proteins involved in the TNFR1 signaling pathway were assessed by western blot. ♦ RESULTS: Incubation with PDF led to a significant up-regulation of TNFR1 on the cell surface correlating with elevated TNFR1 numbers on HPMCs from PD patients. Investigations of underlying molecular mechanisms of TNFR1 signaling showed that PDF affects TNFR1 signaling at the proapoptotic signaling pathway by upregulation of IκBα and downregulation of cFLIPL. In contrast, TNF exclusively induces the activation of NFκB by an increase of phosphorylated IκBα. ♦ CONCLUSIONS: Novel and relevant insights into the mechanisms of TNFR1-mediated signaling in HPMCs with an impact on our understanding of PD-associated damage of the peritoneal membrane are shown.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Inflamación/genética , Epiplón/metabolismo , Diálisis Peritoneal/efectos adversos , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Apoptosis , Western Blotting , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Supervivencia Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/patología , Citometría de Flujo , Humanos , Inflamación/metabolismo , Inflamación/patología , Microscopía Fluorescente , Epiplón/patología , ARN Mensajero/genética , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Transducción de Señal
15.
PLoS One ; 11(10): e0163314, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27755542

RESUMEN

Chronic inflammatory conditions during peritoneal dialysis (PD)-treatment lead to the impairment of peritoneal tissue integrity. The resulting structural and functional reorganization of the peritoneal membrane diminishes ultrafiltration rate and thereby enhances mortality by limiting dialysis effectiveness over time. Tumour necrosis factor (TNF) and its receptors TNFR1 and TNFR2 are key players during inflammatory processes. To date, the role of TNFR1 in peritoneal tissue damage during PD-treatment is completely undefined. In this study, we used an acute PD-mouse model to investigate the role of TNFR1 on structural and morphological changes of the peritoneal membrane. TNFR1-mediated TNF signalling in transgenic mice expressing human TNFR1 was specifically blocked by applying a monoclonal antibody (H398) highly selective for human TNFR1 prior to PD-treatment. Cancer antigen-125 (CA125) plasma concentrations were measured by enzyme-linked immunosorbent assay (ELISA). Western blot analyses were applied to determine TNFR2 protein concentrations. Histological staining of peritoneal tissue sections was performed to assess granulocytes within the peritoneal membrane as well as the content of hyaluronic acid and collagen. We show for the first time that the number of granulocytes within the peritoneal membrane is significantly reduced in mice pre-treated with H398. Moreover, we demonstrate that blocking of TNFR1 not only influences CA125 values but also hyaluronic acid and collagen contents of the peritoneal tissue in these mice. These results strongly suggest that TNFR1 inhibition attenuates peritoneal damage caused by peritoneal dialysis fluid (PDF) and therefore may represent a new therapeutic approach in the treatment of PD-related side effects.


Asunto(s)
Inflamación/prevención & control , Peritoneo/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Antígeno Ca-125/sangre , Colágeno/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Granulocitos/citología , Granulocitos/metabolismo , Ácido Hialurónico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Diálisis Peritoneal , Peritoneo/patología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo
16.
PLoS One ; 8(8): e72156, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977237

RESUMEN

BACKGROUND: Selective inhibition of TNFR1 signaling holds the potential to greatly reduce the pro-inflammatory activity of TNF, while leaving TNFR2 untouched, thus allowing for cell survival and tissue homeostasis. ATROSAB is a humanized antagonistic anti-TNFR1 antibody developed for the treatment of inflammatory diseases. METHODOLOGY/PRINCIPAL FINDINGS: The epitope of ATROSAB resides in the N-terminal region of TNFR1 covering parts of CRD1 and CRD2. By site-directed mutagenesis, we identified Arg68 and His69 of TNFR1 as important residues for ATROSAB binding. ATROSAB inhibited binding of (125)I-labeled TNF to HT1080 in the subnanomolar range. Furthermore, ATROSAB inhibited release of IL-6 and IL-8 from HeLa and HT1080 cells, respectively, induced by TNF or lymphotoxin alpha (LTα). Different from an agonistic antibody (Htr-9), which binds to a region close to the ATROSAB epitope but elicits strong TNFR1 activation, ATROSAB showed a negligible induction of IL-6 and IL-8 production over a broad concentration range. We further verified that ATROSAB, comprising mutations within the Fc region known to abrogate complement fixation and antibody-mediated cellular effector functions, indeed lacks binding activity for C1q, FcγRI (CD64), FcγRIIB (CD32b), and FcγRIII (CD16) disabling ADCC and CDC. CONCLUSIONS/SIGNIFICANCE: The data corroborate ATROSAB's unique function as a TNFR1-selective antagonist efficiently blocking both TNF and LTα action. In agreement with recent studies of TNFR1 complex formation and activation, we suggest a model of the underlying mechanism of TNFR1 inhibition by ATROSAB.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales Humanizados/biosíntesis , Línea Celular Tumoral , Complemento C1q/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Interleucina-8/antagonistas & inhibidores , Interleucina-8/metabolismo , Cinética , Linfotoxina-alfa/farmacología , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Receptores de IgG/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/química , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
17.
Protein Eng Des Sel ; 26(10): 581-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24006371

RESUMEN

IZI-06.1 is a humanized anti-TNFR1 single-chain fragment variable (scFv) that selectively inhibits binding of tumor necrosis factor (TNF) and lymphotoxin alpha to tumor necrosis factor receptor 1 (TNFR1) but not TNFR2. Recently, IZI-06.1 was converted into a fully human IgG1 antibody (ATROSAB) for the treatment of inflammatory diseases. Here, we compare the bivalent ATROSAB with a monovalent scFv-human serum albumin (HSA) fusion protein lacking any antibody-associated effector functions and possessing approximately only half the molecular mass of an IgG, which should facilitate accumulation in inflamed tissues. Furthermore, the half-life of the scFv should be strongly extended while maintaining monovalent binding, avoiding a possible signal transduction by receptor cross-linking in the absence of TNF. The scFv-HSA fusion protein was produced by stably transfected Chinese hamster ovary cells and purified by affinity chromatography. The fusion protein bound specifically to TNFR1 in enzyme-linked immunosorbent assay and TNFR1-transfected mouse embryonic fibroblasts. Affinity determined by quartz crystal microbalance was reduced compared with ATROSAB, which resulted also in a reduced inhibitory activity. Compared with the scFv fragment, the half-life of the fusion protein was significantly increased, although not reaching the long half-life of ATROSAB. In summary, the scFv-HSA may provide an alternative to the full-length IgG1 with the ability to selectively inhibit TNFR1 and exploiting the pharmacokinetic properties of albumin.


Asunto(s)
Ingeniería de Proteínas , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Proteínas Recombinantes de Fusión/genética , Albúmina Sérica/genética , Anticuerpos de Cadena Única/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Células CHO , Cricetinae , Cricetulus , Femenino , Humanos , Ratones , Estabilidad Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Transducción de Señal , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacocinética , Factor de Necrosis Tumoral alfa/metabolismo
18.
Math Biosci Eng ; 9(3): 663-83, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22881031

RESUMEN

Tumor necrosis factor (TNF) is the name giving member of a large cytokine family mirrored by a respective cell membrane receptor super family. TNF itself is a strong proinflammatory regulator of the innate immune system, but has been also recognized as a major factor in progression of autoimmune diseases. A subgroup of the TNF ligand family, including TNF, signals via so-called death receptors, capable to induce a major form of programmed cell death, called apoptosis. Typical for most members of the whole family, death ligands form homotrimeric proteins, capable to bind up to three of their respective receptor molecules. But also unligated receptors occur on the cell surface as homomultimers due to a homophilic interaction domain. Based on these two interaction motivs (ligand/receptor and receptor/receptor) formation of large ligand/receptor clusters can be postulated which have been also observed experimentally. We use here a mass action kinetics approach to establish an ordinary differential equations model describing the dynamics of primary ligand/receptor complex formation as a basis for further clustering on the cell membrane. Based on available experimental data we develop our model in a way that not only ligand/receptor, but also homophilic receptor interaction is encompassed. The model allows formation of two distict primary ligand/receptor complexes in a ligand concentration dependent manner. At extremely high ligand concentrations the system is dominated by ligated receptor homodimers.


Asunto(s)
Modelos Biológicos , Receptores de Muerte Celular/metabolismo , Transducción de Señal/fisiología , Simulación por Computador , Humanos , Cinética , Ligandos , Unión Proteica , Receptores de Muerte Celular/química , Factores de Necrosis Tumoral/química , Factores de Necrosis Tumoral/metabolismo
19.
PLoS One ; 7(8): e42526, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916132

RESUMEN

TNF-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor (TNF) ligand family that exerts its apoptotic activity in human cells by binding to two transmembrane receptors, TRAILR1 and TRAILR2. In cells co-expressing both receptors the particular contribution of either protein to the overall cellular response is not well defined. Here we have investigated whether differences in the signaling capacities of TRAILR1 and TRAILR2 can be attributed to certain functional molecular subdomains. We generated and characterized various chimeric receptors comprising TRAIL receptor domains fused with parts from other members of the TNF death receptor family. This allowed us to compare the contribution of particular domains of the two TRAIL receptors to the overall apoptotic response and to identify elements that regulate apoptotic signaling. Our results show that the TRAIL receptor death domains are weak apoptosis inducers compared to those of CD95/Fas, because TRAILR-derived constructs containing the CD95/Fas death domain possessed strongly enhanced apoptotic capabilities. Importantly, major differences in the signaling strengths of the two TRAIL receptors were linked to their transmembrane domains in combination with the adjacent extracellular stalk regions. This was evident from receptor chimeras comprising the extracellular part of TNFR1 and the intracellular signaling part of CD95/Fas. Both receptor chimeras showed comparable ligand binding affinities and internalization kinetics. However, the respective TRAILR2-derived molecule more efficiently induced apoptosis. It also activated caspase-8 and caspase-3 more strongly and more quickly, albeit being expressed at lower levels. These results suggest that the transmembrane domains together with their adjacent stalk regions can play a major role in control of death receptor activation thereby contributing to cell type specific differences in TRAILR1 and TRAILR2 signaling.


Asunto(s)
Apoptosis , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Transducción de Señal , Animales , Línea Celular Transformada , Endocitosis , Ligandos , Ratones , Unión Proteica , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
20.
EURASIP J Bioinform Syst Biol ; 2012(1): 4, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22651376

RESUMEN

In recent years, cell population models have become increasingly common. In contrast to classic single cell models, population models allow for the study of cell-to-cell variability, a crucial phenomenon in most populations of primary cells, cancer cells, and stem cells. Unfortunately, tools for in-depth analysis of population models are still missing. This problem originates from the complexity of population models. Particularly important are methods to determine the source of heterogeneity (e.g., genetics or epigenetic differences) and to select potential (bio-)markers. We propose an analysis based on visual analytics to tackle this problem. Our approach combines parallel-coordinates plots, used for a visual assessment of the high-dimensional dependencies, and nonlinear support vector machines, for the quantification of effects. The method can be employed to study qualitative and quantitative differences among cells. To illustrate the different components, we perform a case study using the proapoptotic signal transduction pathway involved in cellular apoptosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA