Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 141(2): 315-30, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20403326

RESUMO

RIG-I detects invading viral RNA and activates the transcription factors NF-kappaB and IRF3 through the mitochondrial protein MAVS. Here we show that RNA bearing 5'-triphosphate strongly activates the RIG-I-IRF3 signaling cascade in a reconstituted system composed of RIG-I, mitochondria, and cytosol. Activation of RIG-I requires not only RNA but also polyubiquitin chains linked through lysine 63 (K63) of ubiquitin. RIG-I binds specifically to K63-polyubiquitin chains through its tandem CARD domains in a manner that depends on RNA and ATP. Mutations in the CARD domains that abrogate ubiquitin binding also impair RIG-I activation. Remarkably, unanchored K63-ubiquitin chains, which are not conjugated to any target protein, potently activate RIG-I. These ubiquitin chains function as an endogenous ligand of RIG-I in human cells. Our results delineate the mechanism of RIG-I activation, identify CARD domains as a ubiquitin sensor, and demonstrate that unanchored K63-polyubiquitin chains are signaling molecules in antiviral innate immunity.


Assuntos
RNA Helicases DEAD-box/metabolismo , Imunidade Inata , RNA Viral/imunologia , Transdução de Sinais , Enzimas de Conjugação de Ubiquitina/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/imunologia , Humanos , Quinase I-kappa B/metabolismo , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Polifosfatos/metabolismo , Poliubiquitina/metabolismo , RNA de Cadeia Dupla/imunologia , Receptores Imunológicos , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/metabolismo
2.
Annu Rev Biochem ; 78: 769-96, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19489733

RESUMO

Nuclear factor kappa enhancer binding protein (NF-kappaB) regulates diverse biological processes including immunity, inflammation, and apoptosis. A vast array of cellular stimuli converges on NF-kappaB, and ubiquitination plays an essential role in the coordination of these signals to regulate NF-kappaB activity. At least three steps in NF-kappaB activation directly involve ubiquitination: proteasomal degradation of inhibitor of NF-kappaB (IkappaB), processing of NF-kappaB precursors, and activation of the transforming growth factor (TGF)-beta-activated kinase (TAK1) and IkappaB kinase (IKK) complexes. In this review, we discuss recent advances in the identification and characterization of ubiquitination and deubiquitination machinery that regulate NF-kappaB. Particular emphasis is given to proteasome-independent functions of ubiquitin, specifically its role in the activation of protein kinase complexes and in coordination of cell survival and apoptosis signals downstream of tumor necrosis factor alpha (TNFalpha).


Assuntos
NF-kappa B/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Animais , Apoptose , Sobrevivência Celular , Humanos , Ubiquitinação
3.
Genes Dev ; 31(9): 904-915, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28546513

RESUMO

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.


Assuntos
Proteína Axina/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Proteína Axina/química , Proteína Axina/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Osteossarcoma/genética , Osteossarcoma/metabolismo , Conformação Proteica , Proteólise , Homologia de Sequência , Células Tumorais Cultivadas , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
4.
Mol Cell ; 58(3): 522-33, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25891077

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Oncogênicas/metabolismo , Fosfoproteínas/metabolismo , Receptores Wnt/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Ligação a DNA/genética , Proteínas Desgrenhadas , Citometria de Fluxo , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Immunoblotting , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Microscopia de Fluorescência , Mutação , Proteínas Oncogênicas/genética , Fosfoproteínas/genética , Ligação Proteica , Proteólise , Interferência de RNA , Receptores Wnt/genética , Ubiquitina-Proteína Ligases/genética , Via de Sinalização Wnt/genética
5.
Nonlinear Dyn ; : 1-17, 2023 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-37361002

RESUMO

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.

6.
Immunity ; 36(6): 959-73, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22705106

RESUMO

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.


Assuntos
RNA Helicases DEAD-box/fisiologia , Vírus da Encefalomiocardite/fisiologia , Poliubiquitina/metabolismo , Animais , Sistema Livre de Células , Proteína DEAD-box 58 , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Vírus da Encefalomiocardite/genética , Fibroblastos/metabolismo , Fibroblastos/virologia , Células HEK293/metabolismo , Células HEK293/virologia , Humanos , Fator Regulador 3 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Interferon beta/biossíntese , Interferon beta/genética , Camundongos , Complexos Multiproteicos , Mutagênese Sítio-Dirigida , Ligação Proteica , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , RNA Viral/metabolismo , Receptores Imunológicos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade , Ubiquitinação
7.
Nature ; 517(7533): 223-6, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25327252

RESUMO

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.


Assuntos
Poli Adenosina Difosfato Ribose/metabolismo , Processamento de Proteína Pós-Traducional , Ubiquitina-Proteína Ligases/metabolismo , Adenosina Difosfato Ribose/química , Adenosina Difosfato Ribose/metabolismo , Regulação Alostérica , Animais , Biocatálise , Cristalografia por Raios X , Ativação Enzimática , Humanos , Ligantes , Camundongos , Modelos Moleculares , Poli Adenosina Difosfato Ribose/química , Ligação Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato , Tanquirases/metabolismo , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitinação
8.
Trends Biochem Sci ; 41(9): 773-783, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27377711

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Humanos , Processamento de Proteína Pós-Traducional
9.
Immunity ; 34(3): 285-7, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21435581

RESUMO

Viperin is an interferon-stimulated gene that exerts antiviral effects. In this issue of Immunity, Saitoh et al. (2011) uncovered an unexpected function of Viperin and lipid bodies in interferon induction by Toll-like receptors, specifically in plasmacytoid dendritic cells.

10.
Mol Cell ; 46(6): 735-45, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22705373

RESUMO

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.


Assuntos
GMP Cíclico/análogos & derivados , Imunidade Inata , Proteínas de Membrana/química , Transdução de Sinais/imunologia , Sequência de Aminoácidos , Sítios de Ligação , GMP Cíclico/metabolismo , Dimerização , Humanos , Ligação de Hidrogênio , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Estrutura Terciária de Proteína
11.
Proc Natl Acad Sci U S A ; 110(31): 12649-54, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23847203

RESUMO

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.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mutação , Proteínas Oncogênicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina , Aciltransferases , Antineoplásicos/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Ensaios Clínicos Fase I como Assunto , Proteínas de Ligação a DNA/genética , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Oncogênicas/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Ubiquitina-Proteína Ligases , Proteínas Wnt/genética , Via de Sinalização Wnt
12.
EMBO Rep ; 14(12): 1120-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24165923

RESUMO

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.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Trombospondinas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt , Motivos de Aminoácidos , Sítios de Ligação , Células HEK293 , Humanos , Ligação Proteica , Trombospondinas/química
13.
Nature ; 461(7260): 114-9, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19675569

RESUMO

TRAF6 is a ubiquitin ligase that is essential for the activation of NF-kappaB and MAP kinases in several signalling pathways, including those emanating from the interleukin 1 and Toll-like receptors. TRAF6 functions together with a ubiquitin-conjugating enzyme complex consisting of UBC13 (also known as UBE2N) and UEV1A (UBE2V1) to catalyse Lys 63-linked polyubiquitination, which activates the TAK1 (also known as MAP3K7) kinase complex. TAK1 in turn phosphorylates and activates IkappaB kinase (IKK), leading to the activation of NF-kappaB. Although several proteins are known to be polyubiquitinated in the IL1R and Toll-like receptor pathways, it is not clear whether ubiquitination of any of these proteins is important for TAK1 or IKK activation. By reconstituting TAK1 activation in vitro using purified proteins, here we show that free Lys 63 polyubiquitin chains, which are not conjugated to any target protein, directly activate TAK1 by binding to the ubiquitin receptor TAB2 (also known as MAP3K7IP2). This binding leads to autophosphorylation and activation of TAK1. Furthermore, we found that unanchored polyubiquitin chains synthesized by TRAF6 and UBCH5C (also known as UBE2D3) activate the IKK complex. Disassembly of the polyubiquitin chains by deubiquitination enzymes prevented TAK1 and IKK activation. These results indicate that unanchored polyubiquitin chains directly activate TAK1 and IKK, suggesting a new mechanism of protein kinase regulation.


Assuntos
Quinase I-kappa B/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Poliubiquitina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Enzima Desubiquitinante CYLD , Ativação Enzimática/efeitos dos fármacos , Células HeLa , Humanos , Interleucina-1beta/farmacologia , Lisina/metabolismo , Fosforilação , Poliubiquitina/biossíntese , Receptores Imunológicos , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Enzimas de Conjugação de Ubiquitina , Ubiquitinação
14.
IEEE Trans Cybern ; 52(10): 10276-10289, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33750728

RESUMO

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.


Assuntos
Algoritmos , Redes Neurais de Computação , Aprendizagem , Modelos Teóricos
15.
Immunother Adv ; 2(1): ltac019, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36196369

RESUMO

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.

16.
Sci Rep ; 11(1): 1399, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446805

RESUMO

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.


Assuntos
Imunidade Celular , Proteínas de Neoplasias/imunologia , Neoplasias Experimentais/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Transdução de Sinais/genética
17.
Elife ; 82019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31741433

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Carcinoma Pulmonar de Células não Pequenas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular Tumoral , Proteínas Culina , Receptores ErbB/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Metionil Aminopeptidases/metabolismo , Camundongos , Camundongos Nus , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcriptoma , Ubiquitina-Proteína Ligases/genética , Proteínas de Sinalização YAP , Proteína rhoA de Ligação ao GTP/metabolismo
18.
Nat Commun ; 10(1): 4184, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31519875

RESUMO

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.


Assuntos
Proteína Axina/metabolismo , Peptidase 7 Específica de Ubiquitina/metabolismo , beta Catenina/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Proteína Axina/genética , Linhagem Celular , Linhagem Celular Tumoral , Citometria de Fluxo , Células HCT116 , Humanos , Imunoprecipitação , Camundongos , Osteoblastos/metabolismo , Estabilidade Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peptidase 7 Específica de Ubiquitina/genética , Ubiquitinação/genética , Ubiquitinação/fisiologia , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia , beta Catenina/genética
19.
Elife ; 72018 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-30355448

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Mucosa/metabolismo , Mucosa/patologia , Transdução de Sinais , Actinas/metabolismo , Proteínas de Transporte/química , Membrana Celular/metabolismo , Epitélio/metabolismo , Epitélio/patologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Junções Intercelulares/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Ligação Proteica , Domínios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Ubiquitinação
20.
Oncoimmunology ; 7(2): e1385690, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29308307

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA