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










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(2): e0292655, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38329960

RESUMEN

Thioredoxin-interacting protein (TXNIP) has emerged as a key player in cancer and diabetes since it targets thioredoxin (TRX)-mediated redox regulation and glucose transporter (GLUT)-mediated metabolism. TXNIP consists of two arrestin (ARR, N-ARR and C-ARR) domains at its amino-terminus and two PPxY (PY) motifs and a di-leucine (LL) motif for endocytosis at its carboxyl-terminus. Here, we report that TXNIP shuffles between TRX and GLUTs to regulate homeostasis of intracellular oxidative stress and glucose metabolism. While TXNIP functions as a gatekeeper of TRX by default, it robustly interacted with class I GLUTs through its C-ARR domain upon increase of intracellular reactive oxygen species. This interaction prompted the surface expression downregulation and lysosomal degradation of GLUTs by its carboxyl-terminal LL endocytic signaling motif to attenuate glucose uptake. Consequently, TXNIP expression significantly limited glucose uptake, leading to the suppression of glycolysis, hexosamine biosynthesis, and the pentose phosphate pathway. Our findings establish a fundamental link between ROS and glucose metabolism through TXNIP and provide a promising target for the drug development against GLUT-related metabolic disorders.


Asunto(s)
Proteínas Portadoras , Diabetes Mellitus , Estrés Oxidativo , Tiorredoxinas , Humanos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Glucosa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Animales , Ratones
2.
Nat Cell Biol ; 25(1): 92-107, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36604592

RESUMEN

RIPK3-ZBP1-MLKL-mediated necroptosis is a proinflammatory cell death process that is crucial for antiviral host defence. RIPK3 self-oligomerization and autophosphorylation are prerequisites for executing necroptosis, yet the underlying mechanism of virus-induced RIPK3 activation remains elusive. Interferon-inducible 2'-5' oligoadenylate synthetase-like (OASL) protein is devoid of enzymatic function but displays potent antiviral activity. Here we describe a role of OASL as a virus-induced necroptosis promoter that scaffolds the RIPK3-ZBP1 non-canonical necrosome via liquid-like phase condensation. This liquid-like platform of OASL recruits RIPK3 and ZBP1 via protein-protein interactions to provide spatial segregation for RIPK3 nucleation. This process facilitates the amyloid-like fibril formation and activation of RIPK3 and thereby MLKL phosphorylation for necroptosis. Mice deficient in Oasl1 exhibit severely impaired necroptosis and attenuated inflammation after viral infection, resulting in uncontrolled viral dissemination and lethality. Our study demonstrates an interferon-induced innate response whereby OASL scaffolds RIPK3-ZBP1 assembly via its phase-separated liquid droplets to facilitate necroptosis-mediated antiviral immunity.


Asunto(s)
Necroptosis , Proteínas Quinasas , Animales , Ratones , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Muerte Celular , Antivirales , Interferones/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Apoptosis , Proteínas de Unión al ARN/metabolismo
3.
Res Sq ; 2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-36482980

RESUMEN

Understanding how proteins evolve under selective pressure is a longstanding challenge. The immensity of the search space has limited efforts to systematically evaluate the impact of multiple simultaneous mutations, so mutations have typically been assessed individually. However, epistasis, or the way in which mutations interact, prevents accurate prediction of combinatorial mutations based on measurements of individual mutations. Here, we use artificial intelligence to define the entire functional sequence landscape of a protein binding site in silico, and we call this approach Complete Combinatorial Mutational Enumeration (CCME). By leveraging CCME, we are able to construct a comprehensive map of the evolutionary connectivity within this functional sequence landscape. As a proof of concept, we applied CCME to the ACE2 binding site of the SARS-CoV-2 spike protein receptor binding domain. We selected representative variants from across the functional sequence landscape for testing in the laboratory. We identified variants that retained functionality to bind ACE2 despite changing over 40% of evaluated residue positions, and the variants now escape binding and neutralization by monoclonal antibodies. This work represents a crucial initial stride towards achieving precise predictions of pathogen evolution, opening avenues for proactive mitigation.

4.
Cell Rep ; 39(9): 110904, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35617962

RESUMEN

Despite SARS-CoV-2 being a "novel" virus, early detection of anti-spike IgG in severe COVID-19 patients may be caused by the amplification of humoral memory responses against seasonal coronaviruses. Here, we examine this phenomenon by characterizing anti-spike IgG responses in non-hospitalized convalescent individuals across a spectrum of COVID-19 severity. We observe that disease severity positively correlates with anti-spike IgG levels, IgG cross-reactivity against other betacoronaviruses (ß-CoVs), and FcγR activation. Analysis of IgG targeting ß-CoV-conserved and non-conserved immunodominant epitopes within the SARS-CoV-2 spike protein revealed epitope-specific relationships: IgG targeting the conserved heptad repeat (HR) 2 region significantly correlates with milder disease, while targeting the conserved S2'FP region correlates with more severe disease. Furthermore, a lower HR2-to-S2'FP IgG-binding ratio correlates with greater disease severity, with ICU-hospitalized COVID-19 patients showing the lowest HR2/S2'FP ratios. These findings suggest that HR2/S2'FP IgG profiles may predict disease severity and offer insight into protective versus deleterious humoral recall responses.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Inmunoglobulina G , Estaciones del Año , Glicoproteína de la Espiga del Coronavirus
5.
Nat Rev Microbiol ; 16(6): 341-354, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29556036

RESUMEN

Autophagy is a powerful tool that host cells use to defend against viral infection. Double-membrane vesicles, termed autophagosomes, deliver trapped viral cargo to the lysosome for degradation. Specifically, autophagy initiates an innate immune response by cooperating with pattern recognition receptor signalling to induce interferon production. It also selectively degrades immune components associated with viral particles. Following degradation, autophagy coordinates adaptive immunity by delivering virus-derived antigens for presentation to T lymphocytes. However, in an ongoing evolutionary arms race, viruses have acquired the potent ability to hijack and subvert autophagy for their benefit. In this Review, we focus on the key regulatory steps during viral infection in which autophagy is involved and discuss the specific molecular mechanisms that diverse viruses use to repurpose autophagy for their life cycle and pathogenesis.


Asunto(s)
Autofagia/fisiología , Virosis/inmunología , Virus/inmunología , Animales , Humanos , Inmunidad Innata , Virosis/virología , Virus/metabolismo
6.
Immunity ; 48(3): 474-476, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29562196

RESUMEN

Hosts respond to viral infection by expressing interferon-stimulated genes, of which IFITs are potent inhibitors of viral RNA translation. Johnson et al. (2018) solved the structure of the IFIT1-IFIT3 complex bound cap 0 RNA and explored their concerted antiviral activity.


Asunto(s)
Proteínas Portadoras/genética , ARN Viral , Proteínas Adaptadoras Transductoras de Señales , Síndromes Periódicos Asociados a Criopirina , Humanos , Péptidos y Proteínas de Señalización Intracelular , Biosíntesis de Proteínas , Caperuzas de ARN , Proteínas de Unión al ARN
7.
Nat Microbiol ; 2(11): 1558-1570, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28827581

RESUMEN

Blood CD14+ monocytes are frontline immunomodulators categorized into classical, intermediate or non-classical subsets, and subsequently differentiated into M1 pro- or M2 anti-inflammatory macrophages on stimulation. Although the Zika virus (ZIKV) rapidly establishes viraemia, the target cells and immune responses, particularly during pregnancy, remain elusive. Furthermore, it is unknown whether African- and Asian-lineage ZIKV have different phenotypic impacts on host immune responses. Using human blood infection, we identified CD14+ monocytes as the primary target for African- or Asian-lineage ZIKV infection. When immunoprofiles of human blood infected with ZIKV were compared, a classical/intermediate monocyte-mediated M1-skewed inflammation by the African-lineage ZIKV infection was observed, in contrast to a non-classical monocyte-mediated M2-skewed immunosuppression by the Asian-lineage ZIKV infection. Importantly, infection of the blood of pregnant women revealed an enhanced susceptibility to ZIKV infection. Specifically, Asian-lineage ZIKV infection of pregnant women's blood led to an exacerbated M2-skewed immunosuppression of non-classical monocytes in conjunction with a global suppression of type I interferon-signalling pathway and an aberrant expression of host genes associated with pregnancy complications. Also, 30 ZIKV+ sera from symptomatic pregnant patients showed elevated levels of M2-skewed immunosuppressive cytokines and pregnancy-complication-associated fibronectin-1. This study demonstrates the differential immunomodulatory responses of blood monocytes, particularly during pregnancy, on infection with different lineages of ZIKV.


Asunto(s)
Tolerancia Inmunológica , Receptores de Lipopolisacáridos/inmunología , Monocitos/virología , Complicaciones Infecciosas del Embarazo/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/fisiología , Adolescente , Adulto , Diferenciación Celular , Citocinas/sangre , Citocinas/inmunología , Femenino , Fibronectinas , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Macrófagos/virología , Monocitos/fisiología , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Transducción de Señal , Adulto Joven , Virus Zika/genética , Virus Zika/inmunología , Infección por el Virus Zika/virología
8.
Nat Microbiol ; 2: 17114, 2017 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-28714967

RESUMEN

Many bacteria, including Legionella pneumophila, rely on the type IV secretion system to translocate a repertoire of effector proteins into the hosts for their survival and growth. Type IV coupling protein (T4CP) is a hexameric ATPase that links translocating substrates to the transenvelope secretion conduit. Yet, how a large number of effector proteins are selectively recruited and processed by T4CPs remains enigmatic. DotL, the T4CP of L. pneumophila, contains an ATPase domain and a C-terminal extension whose function is unknown. Unlike T4CPs involved in plasmid DNA translocation, DotL appeared to function by forming a multiprotein complex with four other proteins. Here, we show that the C-terminal extension of DotL interacts with DotN, IcmS, IcmW and an additionally identified subunit LvgA, and that this pentameric assembly binds Legionella effector proteins. We determined the crystal structure of this assembly and built an architecture of the T4CP holocomplex by combining a homology model of the ATPase domain of DotL. The holocomplex is a hexamer of a bipartite structure composed of a membrane-proximal ATPase domain and a membrane-distal substrate-recognition assembly. The presented information demonstrates the architecture and functional dissection of the multiprotein T4CP complexes and provides important insights into their substrate recruitment and processing.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Legionella pneumophila/química , Complejos Multiproteicos/química , Sistemas de Secreción Tipo IV/química , Proteínas Bacterianas/genética , Cristalografía por Rayos X , Regulación Bacteriana de la Expresión Génica , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Dominios Proteicos , Sistemas de Secreción Tipo IV/genética , Sistemas de Secreción Tipo IV/metabolismo
9.
J Exp Med ; 211(7): 1333-47, 2014 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-24958845

RESUMEN

Linear ubiquitination is a newly discovered posttranslational modification that is currently restricted to a small number of known protein substrates. The linear ubiquitination assembly complex (LUBAC), consisting of HOIL-1L, HOIP, and Sharpin, has been reported to activate NF-κB-mediated transcription in response to receptor signaling by ligating linear ubiquitin chains to Nemo and Rip1. Despite recent advances, the detailed roles of LUBAC in immune cells remain elusive. We demonstrate a novel HOIL-1L function as an essential regulator of the activation of the NLRP3/ASC inflammasome in primary bone marrow-derived macrophages (BMDMs) independently of NF-κB activation. Mechanistically, HOIL-1L is required for assembly of the NLRP3/ASC inflammasome and the linear ubiquitination of ASC, which we identify as a novel LUBAC substrate. Consequently, we find that HOIL-1L(-/-) mice have reduced IL-1ß secretion in response to in vivo NLRP3 stimulation and survive lethal challenge with LPS. Together, these data demonstrate that linear ubiquitination is required for NLRP3 inflammasome activation, defining the molecular events of NLRP3 inflammasome activation and expanding the role of LUBAC as an innate immune regulator. Furthermore, our observation is clinically relevant because patients lacking HOIL-1L expression suffer from pyogenic bacterial immunodeficiency, providing a potential new therapeutic target for enhancing inflammation in immunodeficient patients.


Asunto(s)
Proteínas Portadoras/inmunología , Inmunidad Innata/fisiología , Inflamasomas/inmunología , Macrófagos/inmunología , Complejos Multiproteicos/inmunología , Ubiquitinación/fisiología , Animales , Proteínas Portadoras/genética , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/inmunología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Lipopolisacáridos/toxicidad , Macrófagos/citología , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , FN-kappa B/genética , FN-kappa B/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología
10.
Antioxid Redox Signal ; 20(3): 495-506, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-23642014

RESUMEN

SIGNIFICANCE: The autophagy and inflammasome pathways are ancient innate immune mechanisms for controlling invading pathogens that are linked by mutual regulation. In addition to controlling the metabolic homeostasis of the cell through nutrient recycling, the "self-eating" process of autophagy is also responsible for the degradation of damaged organelles, cells, and pathogens to protect the integrity of the organism. As a cytosolic pathogen recognition receptor (PRR) complex, the inflammasome both induces and is induced by autophagy through direct interactions with autophagy proteins or through the effects of secondary molecules, such as mitochondrial reactive oxygen species and mitochondrial DNA. RECENT ADVANCES: While the molecular mechanisms of inflammasome activation and regulation are largely unknown, much of the current knowledge has been established through investigation of the role of autophagy in innate immunity. Likewise, regulatory proteins in the NOD-like receptor family, which includes inflammasome PRRs, are able to stimulate autophagy in response to the presence of a pathogen. CRITICAL ISSUES: Many of the newly uncovered links between autophagy and inflammasomes have raised new questions about the mechanisms controlling inflammasome function, which are highlighted in this review. FUTURE DIRECTIONS: Our basic understanding of the mutual regulation of inflammasomes and autophagy will be essential for designing therapeutics for chronic inflammatory diseases, especially those for which autophagy and inflammasome genes have already been linked.


Asunto(s)
Autofagia/genética , Inmunidad Innata/genética , Inflamasomas/genética , Inflamación/genética , Autofagia/fisiología , Homeostasis/genética , Humanos , Inflamación/inmunología , Inflamación/terapia , Mitocondrias/metabolismo , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...