Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nonlinear Dyn ; : 1-17, 2023 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-37361002

RESUMEN

The COVID-19 pandemic has created an urgent need for mathematical models that can project epidemic trends and evaluate the effectiveness of mitigation strategies. A major challenge in forecasting the transmission of COVID-19 is the accurate assessment of the multiscale human mobility and how it impacts infection through close contacts. By combining the stochastic agent-based modeling strategy and hierarchical structures of spatial containers corresponding to the notion of geographical places, this study proposes a novel model, Mob-Cov, to study the impact of human traveling behavior and individual health conditions on the disease outbreak and the probability of zero-COVID in the population. Specifically, individuals perform power law-type local movements within a container and global transport between different-level containers. It is revealed that frequent long-distance movements inside a small-level container (e.g., a road or a county) and a small population size reduce both the local crowdedness and disease transmission. It takes only half of the time to induce global disease outbreaks when the population increases from 150 to 500 (normalized unit). When the exponent c1 of the long-tail distribution of distance k moved in the same-level container, p(k)∼k-c1·level, increases, the outbreak time decreases rapidly from 75 to 25 (normalized unit). In contrast, travel between large-level containers (e.g., cities and nations) facilitates global spread of the disease and outbreak. When the mean traveling distance across containers 1d increases from 0.5 to 1 (normalized unit), the outbreak occurs almost twice as fast. Moreover, dynamic infection and recovery in the population are able to drive the bifurcation of the system to a "zero-COVID" state or to a "live with COVID" state, depending on the mobility patterns, population number and health conditions. Reducing population size and restricting global travel help achieve zero-COVID-19. Specifically, when c1 is smaller than 0.2, the ratio of people with low levels of mobility is larger than 80% and the population size is smaller than 400, zero-COVID can be achieved within fewer than 1000 time steps. In summary, the Mob-Cov model considers more realistic human mobility at a wide range of spatial scales, and has been designed with equal emphasis on performance, low simulation cost, accuracy, ease of use and flexibility. It is a useful tool for researchers and politicians to apply when investigating pandemic dynamics and when planning actions against disease. Supplementary Information: The online version contains supplementary material available at 10.1007/s11071-023-08489-5.

2.
Immunother Adv ; 2(1): ltac019, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36196369

RESUMEN

Objectives: Sabatolimab is a humanized monoclonal antibody (hIgG4, S228P) directed against human T-cell immunoglobulin domain and mucin domain-3 (TIM-3). Herein, we describe the development and characterization of sabatolimab. Methods: Sabatolimab was tested for binding to its target TIM-3 and blocking properties. The functional effects of sabatolimab were tested in T-cell killing and myeloid cell cytokine assays. Antibody-mediated cell phagocytosis (ADCP) by sabatolimab was also assessed. Results: Sabatolimab was shown to (i) enhance T-cell killing and inflammatory cytokine production by dendritic cells (DCs); (ii) facilitate the phagocytic uptake of TIM-3-expressing target cells; and (iii) block the interaction between TIM-3 and its ligands PtdSer/galectin-9. Conclusion: Taken together, our results support both direct anti-leukemic effects and immune-mediated modulation by sabatolimab, reinforcing the notion that sabatolimab represents a novel immunotherapy with immuno-myeloid activity, holding promise for the treatment of myeloid cell neoplasms.

3.
IEEE Trans Cybern ; 52(10): 10276-10289, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33750728

RESUMEN

Deep kernel learning (DKL) leverages the connection between the Gaussian process (GP) and neural networks (NNs) to build an end-to-end hybrid model. It combines the capability of NN to learn rich representations under massive data and the nonparametric property of GP to achieve automatic regularization that incorporates a tradeoff between model fit and model complexity. However, the deterministic NN encoder may weaken the model regularization of the following GP part, especially on small datasets, due to the free latent representation. We, therefore, present a complete deep latent-variable kernel learning (DLVKL) model wherein the latent variables perform stochastic encoding for regularized representation. We further enhance the DLVKL from two aspects: 1) the expressive variational posterior through neural stochastic differential equation (NSDE) to improve the approximation quality and 2) the hybrid prior taking knowledge from both the SDE prior and the posterior to arrive at a flexible tradeoff. Extensive experiments imply that DLVKL-NSDE performs similar to the well-calibrated GP on small datasets, and shows superiority on large datasets.


Asunto(s)
Algoritmos , Redes Neurales de la Computación , Aprendizaje , Modelos Teóricos
4.
Sci Rep ; 11(1): 1399, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33446805

RESUMEN

SHP2 is a ubiquitous tyrosine phosphatase involved in regulating both tumor and immune cell signaling. In this study, we discovered a novel immune modulatory function of SHP2. Targeting this protein with allosteric SHP2 inhibitors promoted anti-tumor immunity, including enhancing T cell cytotoxic function and immune-mediated tumor regression. Knockout of SHP2 using CRISPR/Cas9 gene editing showed that targeting SHP2 in cancer cells contributes to this immune response. Inhibition of SHP2 activity augmented tumor intrinsic IFNγ signaling resulting in enhanced chemoattractant cytokine release and cytotoxic T cell recruitment, as well as increased expression of MHC Class I and PD-L1 on the cancer cell surface. Furthermore, SHP2 inhibition diminished the differentiation and inhibitory function of immune suppressive myeloid cells in the tumor microenvironment. SHP2 inhibition enhanced responses to anti-PD-1 blockade in syngeneic mouse models. Overall, our study reveals novel functions of SHP2 in tumor immunity and proposes that targeting SHP2 is a promising strategy for cancer immunotherapy.


Asunto(s)
Inmunidad Celular , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Transducción de Señal/genética
5.
Elife ; 82019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31741433

RESUMEN

EGFR-mutant NSCLCs frequently respond to EGFR tyrosine kinase inhibitors (TKIs). However, the responses are not durable, and the magnitude of tumor regression is variable, suggesting the existence of genetic modifiers of EGFR dependency. Here, we applied a genome-wide CRISPR-Cas9 screening to identify genetic determinants of EGFR TKI sensitivity and uncovered putative candidates. We show that knockout of RIC8A, essential for G-alpha protein activation, enhanced EGFR TKI-induced cell death. Mechanistically, we demonstrate that RIC8A is a positive regulator of YAP signaling, activation of which rescued the EGFR TKI sensitizing phenotype resulting from RIC8A knockout. We also show that knockout of ARIH2, or other components in the Cullin-5 E3 complex, conferred resistance to EGFR inhibition, in part by promoting nascent protein synthesis through METAP2. Together, these data uncover a spectrum of previously unidentified regulators of EGFR TKI sensitivity in EGFR-mutant human NSCLC, providing insights into the heterogeneity of EGFR TKI treatment responses.


Asunto(s)
Sistemas CRISPR-Cas , Carcinoma de Pulmón de Células no Pequeñas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Proteínas Cullin , Receptores ErbB/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Factores de Intercambio de Guanina Nucleótido/genética , Células HEK293 , Humanos , Metionil Aminopeptidasas/metabolismo , Ratones , Ratones Desnudos , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcriptoma , Ubiquitina-Proteína Ligasas/genética , Proteínas Señalizadoras YAP , Proteína de Unión al GTP rhoA/metabolismo
6.
Nat Commun ; 10(1): 4184, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519875

RESUMEN

Axin is a key scaffolding protein responsible for the formation of the ß-catenin destruction complex. Stability of Axin protein is regulated by the ubiquitin-proteasome system, and modulation of cellular concentration of Axin protein has a profound effect on Wnt/ß-catenin signaling. Although E3s promoting Axin ubiquitination have been identified, the deubiquitinase responsible for Axin deubiquitination and stabilization remains unknown. Here, we identify USP7 as a potent negative regulator of Wnt/ß-catenin signaling through CRISPR screens. Genetic ablation or pharmacological inhibition of USP7 robustly increases Wnt/ß-catenin signaling in multiple cellular systems. USP7 directly interacts with Axin through its TRAF domain, and promotes deubiquitination and stabilization of Axin. Inhibition of USP7 regulates osteoblast differentiation and adipocyte differentiation through increasing Wnt/ß-catenin signaling. Our study reveals a critical mechanism that prevents excessive degradation of Axin and identifies USP7 as a target for sensitizing cells to Wnt/ß-catenin signaling.


Asunto(s)
Proteína Axina/metabolismo , Peptidasa Específica de Ubiquitina 7/metabolismo , beta Catenina/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Proteína Axina/genética , Línea Celular , Línea Celular Tumoral , Citometría de Flujo , Células HCT116 , Humanos , Inmunoprecipitación , Ratones , Osteoblastos/metabolismo , Estabilidad Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Peptidasa Específica de Ubiquitina 7/genética , Ubiquitinación/genética , Ubiquitinación/fisiología , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , beta Catenina/genética
7.
Elife ; 72018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30355448

RESUMEN

Homeostasis at mucosal surfaces requires cross-talk between the environment and barrier epithelial cells. Disruption of barrier function typifies mucosal disease. Here we elucidate a bifunctional role in coordinating this cross-talk for the inflammatory bowel disease risk-gene INAVA. Both activities require INAVA's DUF3338 domain (renamed CUPID). CUPID stably binds the cytohesin ARF-GEF ARNO to effect lateral membrane F-actin assembly underlying cell-cell junctions and barrier function. Unexpectedly, when bound to CUPID, ARNO affects F-actin dynamics in the absence of its canonical activity as a guanine nucleotide-exchange factor. Upon exposure to IL-1ß, INAVA relocates to form cytosolic puncta, where CUPID amplifies TRAF6-dependent polyubiquitination and inflammatory signaling. In this case, ARNO binding to CUPID negatively-regulates polyubiquitination and the inflammatory response. INAVA and ARNO act similarly in primary human macrophages responding to IL-1ß and to NOD2 agonists. Thus, INAVA-CUPID exhibits dual functions, coordinated directly by ARNO, that bridge epithelial barrier function with extracellular signals and inflammation.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Inflamación/metabolismo , Inflamación/patología , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Transducción de Señal , Actinas/metabolismo , Proteínas Portadoras/química , Membrana Celular/metabolismo , Epitelio/metabolismo , Epitelio/patología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Uniones Intercelulares/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitinación
8.
Oncoimmunology ; 7(2): e1385690, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29308307

RESUMEN

Both in vivo data in preclinical cancer models and in vitro data with T cells from patients with advanced cancer support a role for Tim-3 blockade in promoting effective anti-tumor immunity. Consequently, there is considerable interest in the clinical development of antibody-based therapeutics that target Tim-3 for cancer immunotherapy. A challenge to this clinical development is the fact that several ligands for Tim-3 have been identified: galectin-9, phosphatidylserine, HMGB1, and most recently, CEACAM1. These observations raise the important question of which of these multiple receptor:ligand relationships must be blocked by an anti-Tim-3 antibody in order to achieve therapeutic efficacy. Here, we have examined the properties of anti-murine and anti-human Tim-3 antibodies that have shown functional efficacy and find that all antibodies bind to Tim-3 in a manner that interferes with Tim-3 binding to both phosphatidylserine and CEACAM1. Our data have implications for the understanding of Tim-3 biology and for the screening of anti-Tim-3 antibody candidates that will have functional properties in vivo.

9.
Materials (Basel) ; 10(11)2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29149069

RESUMEN

A kriging modeling method is proposed to conduct the temperature uncertainty analysis of an injection mechanism in squeeze casting. A mathematical model of temperature prediction with multi input and single output is employed to estimate the temperature spatiotemporal distributions of the injection mechanism. The kriging model applies different weights to the independent variables according to spatial location of sample points and their correlation, thus reducing the estimation variance. The predicted value of the kriging model is compared with the sample data at the corresponding position to investigate the influence of the temperature uncertainty of the injection mechanism on the injection process including friction. The results indicate that the significant error is observed at a few sample points in the early injection due to the impact of the uncertainty facts. The variance mean and standard deviation obtained by the model calibrated by experimental samples reduce largely in comparison to those obtained from the initial kriging model. This study indicates that model calibration produces more accurate prediction.

10.
Genes Dev ; 31(9): 904-915, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28546513

RESUMEN

The Wnt/ß-catenin signaling pathway plays essential roles in embryonic development and adult tissue homeostasis. Axin is a concentration-limiting factor responsible for the formation of the ß-catenin destruction complex. Wnt signaling itself promotes the degradation of Axin. However, the underlying molecular mechanism and biological relevance of this targeting of Axin have not been elucidated. Here, we identify SIAH1/2 (SIAH) as the E3 ligase mediating Wnt-induced Axin degradation. SIAH proteins promote the ubiquitination and proteasomal degradation of Axin through interacting with a VxP motif in the GSK3-binding domain of Axin, and this function of SIAH is counteracted by GSK3 binding to Axin. Structural analysis reveals that the Axin segment responsible for SIAH binding is also involved in GSK3 binding but adopts distinct conformations in Axin/SIAH and Axin/GSK3 complexes. Knockout of SIAH1 blocks Wnt-induced Axin ubiquitination and attenuates Wnt-induced ß-catenin stabilization. Our data suggest that Wnt-induced dissociation of the Axin/GSK3 complex allows SIAH to interact with Axin not associated with GSK3 and promote its degradation and that SIAH-mediated Axin degradation represents an important feed-forward mechanism to achieve sustained Wnt/ß-catenin signaling.


Asunto(s)
Proteína Axina/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proteína Axina/química , Proteína Axina/genética , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Osteosarcoma/genética , Osteosarcoma/metabolismo , Conformación Proteica , Proteolisis , Homología de Secuencia , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
11.
Methods Mol Biol ; 1481: 39-48, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27590150

RESUMEN

Wnt pathways are critical for embryonic development and adult tissue homeostasis in all multicellular animals. Many regulatory mechanisms exist to control proper signaling output. Recent studies suggest that cell surface Wnt receptor level is controlled by ubiquitination, and serve as a critical regulatory point of Wnt pathway activity as it determines the responsiveness of cells to Wnt signal. Here, we describe flow cytometry, cell surface protein biotinylation, and immunofluorescence pulse-chase methods to probe the surface expression, ubiquitination, and internalization of the Wnt receptors FZD and LRP6.


Asunto(s)
Desarrollo Embrionario/genética , Biología Molecular/métodos , Receptores Wnt/química , Proteínas Wnt/química , Animales , Biotinilación , Membrana Celular/metabolismo , Femenino , Humanos , Embarazo , Receptores Wnt/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/química , beta Catenina/metabolismo
12.
Trends Biochem Sci ; 41(9): 773-783, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27377711

RESUMEN

Wnt pathways are crucial for embryonic development and adult tissue homeostasis in all multicellular animals. Our understanding of Wnt signaling networks has grown increasingly complex. Recent studies have revealed many regulatory proteins that function at the proximal membrane level to fine-tune signaling output and enhance signaling specificity. These proteins regulate crucial points in Wnt signaling, including post-translational modification of Wnt proteins, regulation of Wnt receptor degradation, internalization of Wnt receptor complex, and specific ligand-receptor complex formation. Such regulators not only provide us with molecular details of Wnt regulation but also serve as potential targets for therapeutic intervention. In this review we highlight new insights into Wnt regulation at the plasma membrane, especially newly identified feedback regulators.


Asunto(s)
Membrana Celular/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Humanos , Procesamiento Proteico-Postraduccional
13.
Cancers (Basel) ; 8(6)2016 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-27338477

RESUMEN

Aberrant activation of the Wnt/ß-catenin pathway is frequently found in various cancers, often through mutations of downstream components. Inhibiting ß-catenin signaling in tumors with downstream pathway mutations remains challenging, due to a lack of favorable targets. On the other hand, targeting upstream components of the Wnt pathway is rather straightforward. However, it is difficult to identify tumors addicted to autocrine or paracrine Wnt signaling. Discovery of the R-spondin-ZNRF3/RNF43 signaling module and its genetic alterations in cancers represents a breakthrough in this area. Membrane E3 ligase ZNRF3 and RNF43 are critical negative feedback regulators of the Wnt pathway, which function through promoting ubiquitination and degradation of Wnt receptors. R-spondin proteins (RSPO1-4) serve as natural antagonists of ZNRF3/RNF43. To maintain strong and sustained Wnt/ß-catenin signaling, cancers need to overcome ZNRF3/RNF43-mediated feedback inhibition. Indeed, mutations of RNF43/ZNRF3 and recurrent translocations of RSPO2/RSPO3 have recently been identified in various cancers. Significantly, genetic alterations in RNF43/ZNRF3/RSPO2/RSPO3 have shown promise as predictive biomarkers in pre-clinical models for the efficacy of upstream Wnt inhibitors. In this review, we will discuss the biology of the R-spondin-ZNRF3/RNF43 signaling module, cancer-associated alterations of this signaling module, and their value as biomarkers to identify Wnt-addicted tumors.

14.
Mol Cell ; 58(3): 522-33, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25891077

RESUMEN

Tumor suppressors ZNRF3 and RNF43 inhibit Wnt signaling through promoting degradation of Wnt coreceptors Frizzled (FZD) and LRP6, and this activity is counteracted by stem cell growth factor R-spondin. The mechanism by which ZNRF3 and RNF43 recognize Wnt receptors remains unclear. Here we uncover an unexpected role of Dishevelled (DVL), a positive Wnt regulator, in promoting Wnt receptor degradation. DVL knockout cells have significantly increased cell surface levels of FZD and LRP6. DVL is required for ZNRF3/RNF43-mediated ubiquitination and degradation of FZD. Physical interaction with DVL is essential for the Wnt inhibitory activity of ZNRF3/RNF43. Binding of FZD through the DEP domain of DVL is required for DVL-mediated downregulation of FZD. Fusion of the DEP domain to ZNRF3/RNF43 overcomes their DVL dependency to downregulate FZD. Our study reveals DVL as a dual function adaptor to recruit negative regulators ZNRF3/RNF43 to Wnt receptors to ensure proper control of pathway activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas/metabolismo , Fosfoproteínas/metabolismo , Receptores Wnt/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Unión al ADN/genética , Proteínas Dishevelled , Citometría de Flujo , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Immunoblotting , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Microscopía Fluorescente , Mutación , Proteínas Oncogénicas/genética , Fosfoproteínas/genética , Unión Proteica , Proteolisis , Interferencia de ARN , Receptores Wnt/genética , Ubiquitina-Proteína Ligasas/genética , Vía de Señalización Wnt/genética
15.
Nature ; 517(7533): 223-6, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25327252

RESUMEN

Protein poly(ADP-ribosyl)ation (PARylation) has a role in diverse cellular processes such as DNA repair, transcription, Wnt signalling, and cell death. Recent studies have shown that PARylation can serve as a signal for the polyubiquitination and degradation of several crucial regulatory proteins, including Axin and 3BP2 (refs 7, 8, 9). The RING-type E3 ubiquitin ligase RNF146 (also known as Iduna) is responsible for PARylation-dependent ubiquitination (PARdU). Here we provide a structural basis for RNF146-catalysed PARdU and how PARdU specificity is achieved. First, we show that iso-ADP-ribose (iso-ADPr), the smallest internal poly(ADP-ribose) (PAR) structural unit, binds between the WWE and RING domains of RNF146 and functions as an allosteric signal that switches the RING domain from a catalytically inactive state to an active one. In the absence of PAR, the RING domain is unable to bind and activate a ubiquitin-conjugating enzyme (E2) efficiently. Binding of PAR or iso-ADPr induces a major conformational change that creates a functional RING structure. Thus, RNF146 represents a new mechanistic class of RING E3 ligases, the activities of which are regulated by non-covalent ligand binding, and that may provide a template for designing inducible protein-degradation systems. Second, we find that RNF146 directly interacts with the PAR polymerase tankyrase (TNKS). Disruption of the RNF146-TNKS interaction inhibits turnover of the substrate Axin in cells. Thus, both substrate PARylation and PARdU are catalysed by enzymes within the same protein complex, and PARdU substrate specificity may be primarily determined by the substrate-TNKS interaction. We propose that the maintenance of unliganded RNF146 in an inactive state may serve to maintain the stability of the RNF146-TNKS complex, which in turn regulates the homeostasis of PARdU activity in the cell.


Asunto(s)
Poli Adenosina Difosfato Ribosa/metabolismo , Procesamiento Proteico-Postraduccional , Ubiquitina-Proteína Ligasas/metabolismo , Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Regulación Alostérica , Animales , Biocatálisis , Cristalografía por Rayos X , Activación Enzimática , Humanos , Ligandos , Ratones , Modelos Moleculares , Poli Adenosina Difosfato Ribosa/química , Unión Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato , Tanquirasas/metabolismo , Enzimas Ubiquitina-Conjugadoras/química , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitinación
16.
EMBO Rep ; 14(12): 1120-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24165923

RESUMEN

R-spondin proteins sensitize cells to Wnt signalling and act as potent stem cell growth factors. Various membrane proteins have been proposed as potential receptors of R-spondin, including LGR4/5, membrane E3 ubiquitin ligases ZNRF3/RNF43 and several others proteins. Here, we show that R-spondin interacts with ZNRF3/RNF43 and LGR4 through distinct motifs. Both LGR4 and ZNRF3 binding motifs are required for R-spondin-induced LGR4/ZNRF3 interaction, membrane clearance of ZNRF3 and activation of Wnt signalling. Importantly, Wnt-inhibitory activity of ZNRF3, but not of a ZNRF3 mutant with reduced affinity to R-spondin, can be strongly suppressed by R-spondin, suggesting that R-spondin primarily functions by binding and inhibiting ZNRF3. Together, our results support a dual receptor model of R-spondin action, where LGR4/5 serve as the engagement receptor whereas ZNRF3/RNF43 function as the effector receptor.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Trombospondinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt , Secuencias de Aminoácidos , Sitios de Unión , Células HEK293 , Humanos , Unión Proteica , Trombospondinas/química
17.
Proc Natl Acad Sci U S A ; 110(31): 12649-54, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23847203

RESUMEN

A growing number of agents targeting ligand-induced Wnt/ß-catenin signaling are being developed for cancer therapy. However, clinical development of these molecules is challenging because of the lack of a genetic strategy to identify human tumors dependent on ligand-induced Wnt/ß-catenin signaling. Ubiquitin E3 ligase ring finger 43 (RNF43) has been suggested as a negative regulator of Wnt signaling, and mutations of RNF43 have been identified in various tumors, including cystic pancreatic tumors. However, loss of function study of RNF43 in cell culture has not been conducted, and the functional significance of RNF43 mutations in cancer is unknown. Here, we show that RNF43 inhibits Wnt/ß-catenin signaling by reducing the membrane level of Frizzled in pancreatic cancer cells, serving as a negative feedback mechanism. Inhibition of endogenous Wnt/ß-catenin signaling increased the cell surface level of Frizzled. A panel of 39 pancreatic cancer cell lines was tested for Wnt dependency using LGK974, a selective Porcupine inhibitor being examined in a phase 1 clinical trial. Strikingly, all LGK974-sensitive lines carried inactivating mutations of RNF43. Inhibition of Wnt secretion, depletion of ß-catenin, or expression of wild-type RNF43 blocked proliferation of RNF43 mutant but not RNF43-wild-type pancreatic cancer cells. LGK974 inhibited proliferation and induced differentiation of RNF43-mutant pancreatic adenocarcinoma xenograft models. Our data suggest that mutational inactivation of RNF43 in pancreatic adenocarcinoma confers Wnt dependency, and the presence of RNF43 mutations could be used as a predictive biomarker for patient selection supporting the clinical development of Wnt inhibitors in subtypes of cancer.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas de Unión al ADN/metabolismo , Mutación , Proteínas Oncogénicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina , Aciltransferasas , Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Ensayos Clínicos Fase I como Asunto , Proteínas de Unión al ADN/genética , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Oncogénicas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Ubiquitina-Proteína Ligasas , Proteínas Wnt/genética , Vía de Señalización Wnt
18.
Immunity ; 36(6): 959-73, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22705106

RESUMEN

RIG-I and MDA5 detect viral RNA in the cytoplasm and activate signaling cascades leading to the production of type-I interferons. RIG-I is activated through sequential binding of viral RNA and unanchored lysine-63 (K63) polyubiquitin chains, but how polyubiquitin activates RIG-I and whether MDA5 is activated through a similar mechanism remain unresolved. Here, we showed that the CARD domains of MDA5 bound to K63 polyubiquitin and that this binding was essential for MDA5 to activate the transcription factor IRF3. Mutations of conserved residues in MDA5 and RIG-I that disrupt their ubiquitin binding also abrogated their ability to activate IRF3. Polyubiquitin binding induced the formation of a large complex consisting of four RIG-I and four ubiquitin chains. This hetero-tetrameric complex was highly potent in activating the antiviral signaling cascades. These results suggest a unified mechanism of RIG-I and MDA5 activation and reveal a unique mechanism by which ubiquitin regulates cell signaling and immune response.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Virus de la Encefalomiocarditis/fisiología , Poliubiquitina/metabolismo , Animales , Sistema Libre de Células , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , Virus de la Encefalomiocarditis/genética , Fibroblastos/metabolismo , Fibroblastos/virología , Células HEK293/metabolismo , Células HEK293/virología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferón beta/biosíntesis , Interferón beta/genética , Ratones , Complejos Multiproteicos , Mutagénesis Sitio-Dirigida , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , ARN Viral/metabolismo , Receptores Inmunológicos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/fisiología , Relación Estructura-Actividad , Ubiquitinación
19.
Mol Cell ; 46(6): 735-45, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22705373

RESUMEN

Detection of foreign materials is the first step of successful immune responses. Stimulator of interferon genes (STING) was shown to directly bind cyclic diguanylate monophosphate (c-di-GMP), a bacterial second messenger, and to elicit strong interferon responses. Here we elucidate the structural features of the cytosolic c-di-GMP binding domain (CBD) of STING and its complex with c-di-GMP. The CBD exhibits an α + ß fold and is a dimer in the crystal and in solution. Surprisingly, one c-di-GMP molecule binds to the central crevice of a STING dimer, using a series of stacking and hydrogen bonding interactions. We show that STING is autoinhibited by an intramolecular interaction between the CBD and the C-terminal tail (CTT) and that c-di-GMP releases STING from this autoinhibition by displacing the CTT. The structures provide a remarkable example of pathogen-host interactions in which a unique microbial molecule directly engages the innate immune system.


Asunto(s)
GMP Cíclico/análogos & derivados , Inmunidad Innata , Proteínas de la Membrana/química , Transducción de Señal/inmunología , Secuencia de Aminoácidos , Sitios de Unión , GMP Cíclico/metabolismo , Dimerización , Humanos , Enlace de Hidrógeno , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Estructura Terciaria de Proteína
20.
Nat Rev Immunol ; 12(1): 35-48, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22158412

RESUMEN

Ubiquitylation is a widely used post-translational protein modification that regulates many biological processes, including immune responses. The role of ubiquitin in immune regulation was originally uncovered through studies of antigen presentation and the nuclear factor-κB family of transcription factors, which orchestrate host defence against microorganisms. Recent studies have revealed crucial roles of ubiquitylation in many aspects of the immune system, including innate and adaptive immunity and antimicrobial autophagy. In addition, mounting evidence indicates that microbial pathogens exploit the ubiquitin pathway to evade the host immune system. Here, we review recent advances on the role of ubiquitylation in host defence and pathogen evasion.


Asunto(s)
Inmunidad Adaptativa , Evasión Inmune , Inmunidad Innata , Ubiquitinación/inmunología , Animales , Presentación de Antígeno , Autofagia/inmunología , Humanos , Procesamiento Proteico-Postraduccional/inmunología , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...