Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 828
Filtrar
1.
Annu Rev Immunol ; 42(1): 615-645, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38941608

RESUMEN

The COVID-19 pandemic was caused by the recently emerged ß-coronavirus SARS-CoV-2. SARS-CoV-2 has had a catastrophic impact, resulting in nearly 7 million fatalities worldwide to date. The innate immune system is the first line of defense against infections, including the detection and response to SARS-CoV-2. Here, we discuss the innate immune mechanisms that sense coronaviruses, with a focus on SARS-CoV-2 infection and how these protective responses can become detrimental in severe cases of COVID-19, contributing to cytokine storm, inflammation, long-COVID, and other complications. We also highlight the complex cross talk among cytokines and the cellular components of the innate immune system, which can aid in viral clearance but also contribute to inflammatory cell death, cytokine storm, and organ damage in severe COVID-19 pathogenesis. Furthermore, we discuss how SARS-CoV-2 evades key protective innate immune mechanisms to enhance its virulence and pathogenicity, as well as how innate immunity can be therapeutically targeted as part of the vaccination and treatment strategy. Overall, we highlight how a comprehensive understanding of innate immune mechanisms has been crucial in the fight against SARS-CoV-2 infections and the development of novel host-directed immunotherapeutic strategies for various diseases.


Asunto(s)
COVID-19 , Inmunidad Innata , SARS-CoV-2 , Humanos , COVID-19/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/metabolismo , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/prevención & control , Evasión Inmune
2.
Cell ; 187(15): 4061-4077.e17, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38878777

RESUMEN

NLRs constitute a large, highly conserved family of cytosolic pattern recognition receptors that are central to health and disease, making them key therapeutic targets. NLRC5 is an enigmatic NLR with mutations associated with inflammatory and infectious diseases, but little is known about its function as an innate immune sensor and cell death regulator. Therefore, we screened for NLRC5's role in response to infections, PAMPs, DAMPs, and cytokines. We identified that NLRC5 acts as an innate immune sensor to drive inflammatory cell death, PANoptosis, in response to specific ligands, including PAMP/heme and heme/cytokine combinations. NLRC5 interacted with NLRP12 and PANoptosome components to form a cell death complex, suggesting an NLR network forms similar to those in plants. Mechanistically, TLR signaling and NAD+ levels regulated NLRC5 expression and ROS production to control cell death. Furthermore, NLRC5-deficient mice were protected in hemolytic and inflammatory models, suggesting that NLRC5 could be a potential therapeutic target.


Asunto(s)
Inflamación , Péptidos y Proteínas de Señalización Intracelular , NAD , Animales , Ratones , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , NAD/metabolismo , Humanos , Inmunidad Innata , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Ratones Noqueados , Transducción de Señal , Células HEK293 , Inflamasomas/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Receptores Toll-Like/metabolismo , Masculino , Citocinas/metabolismo , Proteínas de Unión al Calcio
3.
Cell ; 186(13): 2783-2801.e20, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37267949

RESUMEN

Cytosolic innate immune sensors are critical for host defense and form complexes, such as inflammasomes and PANoptosomes, that induce inflammatory cell death. The sensor NLRP12 is associated with infectious and inflammatory diseases, but its activating triggers and roles in cell death and inflammation remain unclear. Here, we discovered that NLRP12 drives inflammasome and PANoptosome activation, cell death, and inflammation in response to heme plus PAMPs or TNF. TLR2/4-mediated signaling through IRF1 induced Nlrp12 expression, which led to inflammasome formation to induce maturation of IL-1ß and IL-18. The inflammasome also served as an integral component of a larger NLRP12-PANoptosome that drove inflammatory cell death through caspase-8/RIPK3. Deletion of Nlrp12 protected mice from acute kidney injury and lethality in a hemolytic model. Overall, we identified NLRP12 as an essential cytosolic sensor for heme plus PAMPs-mediated PANoptosis, inflammation, and pathology, suggesting that NLRP12 and molecules in this pathway are potential drug targets for hemolytic and inflammatory diseases.


Asunto(s)
Inflamasomas , Moléculas de Patrón Molecular Asociado a Patógenos , Animales , Ratones , Inflamasomas/metabolismo , Hemo , Inflamación , Piroptosis , Péptidos y Proteínas de Señalización Intracelular
4.
Annu Rev Immunol ; 33: 79-106, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25493335

RESUMEN

Cell proliferation and cell death are integral elements in maintaining homeostatic balance in metazoans. Disease pathologies ensue when these processes are disturbed. A plethora of evidence indicates that malfunction of cell death can lead to inflammation, autoimmunity, or immunodeficiency. Programmed necrosis or necroptosis is a form of nonapoptotic cell death driven by the receptor interacting protein kinase 3 (RIPK3) and its substrate, mixed lineage kinase domain-like (MLKL). RIPK3 partners with its upstream adaptors RIPK1, TRIF, or DAI to signal for necroptosis in response to death receptor or Toll-like receptor stimulation, pathogen infection, or sterile cell injury. Necroptosis promotes inflammation through leakage of cellular contents from damaged plasma membranes. Intriguingly, many of the signal adaptors of necroptosis have dual functions in innate immune signaling. This unique signature illustrates the cooperative nature of necroptosis and innate inflammatory signaling pathways in managing cell and organismal stresses from pathogen infection and sterile tissue injury.


Asunto(s)
Inflamación/metabolismo , Inflamación/patología , Necrosis/metabolismo , Transducción de Señal , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Evolución Biológica , Muerte Celular , Humanos , Inflamasomas/metabolismo , Inflamación/genética , Interleucina-1beta/metabolismo , FN-kappa B/metabolismo , Enfermedades Parasitarias/genética , Enfermedades Parasitarias/metabolismo , Enfermedades Parasitarias/patología , Fosforilación , Unión Proteica , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ubiquitinación , Virosis/genética , Virosis/metabolismo , Virosis/patología
5.
Cell ; 185(17): 3214-3231.e23, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35907404

RESUMEN

Although mutations in mitochondrial-associated genes are linked to inflammation and susceptibility to infection, their mechanistic contributions to immune outcomes remain ill-defined. We discovered that the disease-associated gain-of-function allele Lrrk2G2019S (leucine-rich repeat kinase 2) perturbs mitochondrial homeostasis and reprograms cell death pathways in macrophages. When the inflammasome is activated in Lrrk2G2019S macrophages, elevated mitochondrial ROS (mtROS) directs association of the pore-forming protein gasdermin D (GSDMD) to mitochondrial membranes. Mitochondrial GSDMD pore formation then releases mtROS, promoting a switch to RIPK1/RIPK3/MLKL-dependent necroptosis. Consistent with enhanced necroptosis, infection of Lrrk2G2019S mice with Mycobacterium tuberculosis elicits hyperinflammation and severe immunopathology. Our findings suggest a pivotal role for GSDMD as an executer of multiple cell death pathways and demonstrate that mitochondrial dysfunction can direct immune outcomes via cell death modality switching. This work provides insights into how LRRK2 mutations manifest or exacerbate human diseases and identifies GSDMD-dependent necroptosis as a potential target to limit Lrrk2G2019S-mediated immunopathology.


Asunto(s)
Mitocondrias , Necroptosis , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Animales , Humanos , Inflamasomas , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Macrófagos , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
6.
Cell ; 184(17): 4447-4463.e20, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34363755

RESUMEN

TANK binding kinase 1 (TBK1) regulates IFN-I, NF-κB, and TNF-induced RIPK1-dependent cell death (RCD). In mice, biallelic loss of TBK1 is embryonically lethal. We discovered four humans, ages 32, 26, 7, and 8 from three unrelated consanguineous families with homozygous loss-of-function mutations in TBK1. All four patients suffer from chronic and systemic autoinflammation, but not severe viral infections. We demonstrate that TBK1 loss results in hypomorphic but sufficient IFN-I induction via RIG-I/MDA5, while the system retains near intact IL-6 induction through NF-κB. Autoinflammation is driven by TNF-induced RCD as patient-derived fibroblasts experienced higher rates of necroptosis in vitro, and CC3 was elevated in peripheral blood ex vivo. Treatment with anti-TNF dampened the baseline circulating inflammatory profile and ameliorated the clinical condition in vivo. These findings highlight the plasticity of the IFN-I response and underscore a cardinal role for TBK1 in the regulation of RCD.


Asunto(s)
Inflamación/enzimología , Proteínas Serina-Treonina Quinasas/deficiencia , Factor de Necrosis Tumoral alfa/farmacología , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Autoinmunidad/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Enzima Desubiquitinante CYLD/metabolismo , Femenino , Células HEK293 , Homocigoto , Humanos , Quinasa I-kappa B/metabolismo , Inmunofenotipificación , Inflamación/patología , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Mutación con Pérdida de Función/genética , Masculino , Linaje , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Receptor Toll-Like 3/metabolismo , Transcriptoma/genética , Vesiculovirus/efectos de los fármacos , Vesiculovirus/fisiología
7.
Cell ; 181(3): 674-687.e13, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32298652

RESUMEN

Caspases regulate cell death, immune responses, and homeostasis. Caspase-6 is categorized as an executioner caspase but shows key differences from the other executioners. Overall, little is known about the functions of caspase-6 in biological processes apart from apoptosis. Here, we show that caspase-6 mediates innate immunity and inflammasome activation. Furthermore, we demonstrate that caspase-6 promotes the activation of programmed cell death pathways including pyroptosis, apoptosis, and necroptosis (PANoptosis) and plays an essential role in host defense against influenza A virus (IAV) infection. In addition, caspase-6 promoted the differentiation of alternatively activated macrophages (AAMs). Caspase-6 facilitated the RIP homotypic interaction motif (RHIM)-dependent binding of RIPK3 to ZBP1 via its interaction with RIPK3. Altogether, our findings reveal a vital role for caspase-6 in facilitating ZBP1-mediated inflammasome activation, cell death, and host defense during IAV infection, opening additional avenues for treatment of infectious and autoinflammatory diseases and cancer.


Asunto(s)
Caspasa 6/inmunología , Caspasa 6/metabolismo , Inflamasomas/inmunología , Animales , Apoptosis/inmunología , Muerte Celular/inmunología , Inmunidad Innata , Inflamasomas/metabolismo , Inflamasomas/fisiología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Necroptosis/inmunología , Unión Proteica , Piroptosis/inmunología , Proteínas de Unión al ARN/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
8.
Cell ; 180(6): 1115-1129.e13, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32200799

RESUMEN

Influenza A virus (IAV) is a lytic RNA virus that triggers receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-mediated pathways of apoptosis and mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptosis in infected cells. ZBP1 initiates RIPK3-driven cell death by sensing IAV RNA and activating RIPK3. Here, we show that replicating IAV generates Z-RNAs, which activate ZBP1 in the nucleus of infected cells. ZBP1 then initiates RIPK3-mediated MLKL activation in the nucleus, resulting in nuclear envelope disruption, leakage of DNA into the cytosol, and eventual necroptosis. Cell death induced by nuclear MLKL was a potent activator of neutrophils, a cell type known to drive inflammatory pathology in virulent IAV disease. Consequently, MLKL-deficient mice manifest reduced nuclear disruption of lung epithelia, decreased neutrophil recruitment into infected lungs, and increased survival following a lethal dose of IAV. These results implicate Z-RNA as a new pathogen-associated molecular pattern and describe a ZBP1-initiated nucleus-to-plasma membrane "inside-out" death pathway with potentially pathogenic consequences in severe cases of influenza.


Asunto(s)
Virus de la Influenza A/genética , Necroptosis/genética , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis/genética , Muerte Celular/genética , Línea Celular Tumoral , Femenino , Virus de la Influenza A/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , ARN/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología
9.
Immunity ; 57(7): 1514-1532.e15, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38788712

RESUMEN

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.


Asunto(s)
Muerte Celular Inmunogénica , Proteolisis , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Humanos , Animales , Ratones , Proteolisis/efectos de los fármacos , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Muerte Celular Inmunogénica/efectos de los fármacos , Necroptosis/efectos de los fármacos , Necroptosis/inmunología , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Ratones Endogámicos C57BL , Antineoplásicos/farmacología , Inmunoterapia/métodos
10.
Immunity ; 57(7): 1497-1513.e6, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38744293

RESUMEN

RIPK1 is a multi-functional kinase that regulates cell death and inflammation and has been implicated in the pathogenesis of inflammatory diseases. RIPK1 acts in a kinase-dependent and kinase-independent manner to promote or suppress apoptosis and necroptosis, but the underlying mechanisms remain poorly understood. Here, we show that a mutation (R588E) disrupting the RIPK1 death domain (DD) caused perinatal lethality induced by ZBP1-mediated necroptosis. Additionally, these mice developed postnatal inflammatory pathology, which was mediated by necroptosis-independent TNFR1, TRADD, and TRIF signaling, partially requiring RIPK3. Our biochemical mechanistic studies revealed that ZBP1- and TRIF-mediated activation of RIPK3 required RIPK1 kinase activity in wild-type cells but not in Ripk1R588E/R588E cells, suggesting that DD-dependent oligomerization of RIPK1 and its interaction with FADD determine the mechanisms of RIPK3 activation by ZBP1 and TRIF. Collectively, these findings revealed a critical physiological role of DD-dependent RIPK1 signaling that is important for the regulation of tissue homeostasis and inflammation.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Inflamación , Necroptosis , Proteínas de Unión al ARN , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Transducción de Señal , Animales , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Ratones , Inflamación/metabolismo , Inflamación/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/genética , Muerte Celular , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Dominios Proteicos , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Apoptosis , Mutación , Proteína de Dominio de Muerte Asociada a Receptor de TNF
11.
Immunity ; 57(3): 429-445, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38479360

RESUMEN

Diverse inflammatory conditions, from infections to autoimmune disease, are often associated with cellular damage and death. Apoptotic cell death has evolved to minimize its inflammatory potential. By contrast, necrotic cell death via necroptosis and pyroptosis-driven by membrane-damaging MLKL and gasdermins, respectively-can both initiate and propagate inflammatory responses. In this review, we provide insights into the function and regulation of MLKL and gasdermin necrotic effector proteins and drivers of plasma membrane rupture. We evaluate genetic evidence that MLKL- and gasdermin-driven necrosis may either provide protection against, or contribute to, disease states in a context-dependent manner. These cumulative insights using gene-targeted mice underscore the necessity for future research examining pyroptotic and necroptotic cell death in human tissue, as a basis for developing specific necrotic inhibitors with the potential to benefit a spectrum of pathological conditions.


Asunto(s)
Apoptosis , Gasderminas , Humanos , Animales , Ratones , Necrosis/metabolismo , Apoptosis/fisiología , Piroptosis/fisiología , Muerte Celular , Inflamasomas/metabolismo , Proteínas Quinasas/metabolismo
12.
Immunity ; 57(4): 674-699, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599165

RESUMEN

Nucleotide-binding oligomerization domain (NOD)-like receptors, also known as nucleotide-binding leucine-rich repeat receptors (NLRs), are a family of cytosolic pattern recognition receptors that detect a wide variety of pathogenic and sterile triggers. Activation of specific NLRs initiates pro- or anti-inflammatory signaling cascades and the formation of inflammasomes-multi-protein complexes that induce caspase-1 activation to drive inflammatory cytokine maturation and lytic cell death, pyroptosis. Certain NLRs and inflammasomes act as integral components of larger cell death complexes-PANoptosomes-driving another form of lytic cell death, PANoptosis. Here, we review the current understanding of the evolution, structure, and function of NLRs in health and disease. We discuss the concept of NLR networks and their roles in driving cell death and immunity. An improved mechanistic understanding of NLRs may provide therapeutic strategies applicable across infectious and inflammatory diseases and in cancer.


Asunto(s)
Inflamasomas , Receptores de Reconocimiento de Patrones , Inflamasomas/metabolismo , Piroptosis , Inmunidad Innata , Nucleótidos
13.
Cell ; 174(6): 1477-1491.e19, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30146158

RESUMEN

Aging is a major risk factor for both genetic and sporadic neurodegenerative disorders. However, it is unclear how aging interacts with genetic predispositions to promote neurodegeneration. Here, we investigate how partial loss of function of TBK1, a major genetic cause for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) comorbidity, leads to age-dependent neurodegeneration. We show that TBK1 is an endogenous inhibitor of RIPK1 and the embryonic lethality of Tbk1-/- mice is dependent on RIPK1 kinase activity. In aging human brains, another endogenous RIPK1 inhibitor, TAK1, exhibits a marked decrease in expression. We show that in Tbk1+/- mice, the reduced myeloid TAK1 expression promotes all the key hallmarks of ALS/FTD, including neuroinflammation, TDP-43 aggregation, axonal degeneration, neuronal loss, and behavior deficits, which are blocked upon inhibition of RIPK1. Thus, aging facilitates RIPK1 activation by reducing TAK1 expression, which cooperates with genetic risk factors to promote the onset of ALS/FTD.


Asunto(s)
Apoptosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Adulto , Anciano , Envejecimiento , Animales , Apoptosis/efectos de los fármacos , Axones/metabolismo , Conducta Animal , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Humanos , Quinasa I-kappa B/metabolismo , Ratones , Ratones Noqueados , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Médula Espinal/metabolismo , Estaurosporina/farmacología , Factor de Necrosis Tumoral alfa/farmacología
14.
Cell ; 173(5): 1244-1253.e10, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29681455

RESUMEN

The RIPK1-RIPK3 necrosome is an amyloid signaling complex that initiates TNF-induced necroptosis, serving in human immune defense, cancer, and neurodegenerative diseases. RIPK1 and RIPK3 associate through their RIP homotypic interaction motifs with consensus sequences IQIG (RIPK1) and VQVG (RIPK3). Using solid-state nuclear magnetic resonance, we determined the high-resolution structure of the RIPK1-RIPK3 core. RIPK1 and RIPK3 alternately stack (RIPK1, RIPK3, RIPK1, RIPK3, etc.) to form heterotypic ß sheets. Two such ß sheets bind together along a compact hydrophobic interface featuring an unusual ladder of alternating Ser (from RIPK1) and Cys (from RIPK3). The crystal structure of a four-residue RIPK3 consensus sequence is consistent with the architecture determined by NMR. The RIPK1-RIPK3 core is the first detailed structure of a hetero-amyloid and provides a potential explanation for the specificity of hetero- over homo-amyloid formation and a structural basis for understanding the mechanisms of signal transduction.


Asunto(s)
Amiloide/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Resonancia Magnética Nuclear Biomolecular , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Alineación de Secuencia
15.
Immunity ; 56(7): 1578-1595.e8, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37329888

RESUMEN

It is currently not well known how necroptosis and necroptosis responses manifest in vivo. Here, we uncovered a molecular switch facilitating reprogramming between two alternative modes of necroptosis signaling in hepatocytes, fundamentally affecting immune responses and hepatocarcinogenesis. Concomitant necrosome and NF-κB activation in hepatocytes, which physiologically express low concentrations of receptor-interacting kinase 3 (RIPK3), did not lead to immediate cell death but forced them into a prolonged "sublethal" state with leaky membranes, functioning as secretory cells that released specific chemokines including CCL20 and MCP-1. This triggered hepatic cell proliferation as well as activation of procarcinogenic monocyte-derived macrophage cell clusters, contributing to hepatocarcinogenesis. In contrast, necrosome activation in hepatocytes with inactive NF-κB-signaling caused an accelerated execution of necroptosis, limiting alarmin release, and thereby preventing inflammation and hepatocarcinogenesis. Consistently, intratumoral NF-κB-necroptosis signatures were associated with poor prognosis in human hepatocarcinogenesis. Therefore, pharmacological reprogramming between these distinct forms of necroptosis may represent a promising strategy against hepatocellular carcinoma.


Asunto(s)
Neoplasias Hepáticas , FN-kappa B , Humanos , FN-kappa B/metabolismo , Proteínas Quinasas/metabolismo , Necroptosis , Inflamación/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Apoptosis
16.
Cell ; 169(2): 301-313.e11, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28366204

RESUMEN

Receptor-interacting protein kinase-3 (RIPK3) is an activator of necroptotic cell death, but recent work has implicated additional roles for RIPK3 in inflammatory signaling independent of cell death. However, while necroptosis has been shown to contribute to antiviral immunity, death-independent roles for RIPK3 in host defense have not been demonstrated. Using a mouse model of West Nile virus (WNV) encephalitis, we show that RIPK3 restricts WNV pathogenesis independently of cell death. Ripk3-/- mice exhibited enhanced mortality compared to wild-type (WT) controls, while mice lacking the necroptotic effector MLKL, or both MLKL and caspase-8, were unaffected. The enhanced susceptibility of Ripk3-/- mice arose from suppressed neuronal chemokine expression and decreased central nervous system (CNS) recruitment of T lymphocytes and inflammatory myeloid cells, while peripheral immunity remained intact. These data identify pleiotropic functions for RIPK3 in the restriction of viral pathogenesis and implicate RIPK3 as a key coordinator of immune responses within the CNS.


Asunto(s)
Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología , Animales , Sistema Nervioso Central/metabolismo , Quimiocinas/inmunología , Leucocitos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Necrosis , Neuronas/metabolismo
17.
Mol Cell ; 84(5): 938-954.e8, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38272024

RESUMEN

Phase separation is a vital mechanism that mediates the formation of biomolecular condensates and their functions. Necroptosis is a lytic form of programmed cell death mediated by RIPK1, RIPK3, and MLKL downstream of TNFR1 and has been implicated in mediating many human diseases. However, whether necroptosis is regulated by phase separation is not yet known. Here, we show that upon the induction of necroptosis and recruitment by the adaptor protein TAX1BP1, PARP5A and its binding partner RNF146 form liquid-like condensates by multivalent interactions to perform poly ADP-ribosylation (PARylation) and PARylation-dependent ubiquitination (PARdU) of activated RIPK1 in mouse embryonic fibroblasts. We show that PARdU predominantly occurs on the K376 residue of mouse RIPK1, which promotes proteasomal degradation of kinase-activated RIPK1 to restrain necroptosis. Our data demonstrate that PARdU on K376 of mouse RIPK1 provides an alternative cell death checkpoint mediated by phase separation-dependent control of necroptosis by PARP5A and RNF146.


Asunto(s)
Necroptosis , Separación de Fases , Animales , Ratones , Apoptosis/fisiología , Muerte Celular , Fibroblastos/metabolismo , Necroptosis/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
18.
Annu Rev Biochem ; 85: 743-63, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26865533

RESUMEN

Necroptosis is a regulated form of necrosis, with the dying cell rupturing and releasing intracellular components that can trigger an innate immune response. Toll-like receptor 3 and 4 agonists, tumor necrosis factor, certain viral infections, or the T cell receptor can trigger necroptosis if the activity of the protease caspase-8 is compromised. Necroptosis signaling is modulated by the kinase RIPK1 and requires the kinase RIPK3 and the pseudokinase MLKL. Either RIPK3 deficiency or RIPK1 inhibition confers resistance in various animal disease models, suggesting that inflammation caused by necroptosis contributes to tissue damage and that inhibitors of these kinases could have therapeutic potential. Recent studies have revealed unexpected complexity in the regulation of cell death programs by RIPK1 and RIPK3 with the possibility that necroptosis is but one mechanism by which these kinases promote inflammation.


Asunto(s)
Regulación de la Expresión Génica , Necrosis/genética , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Animales , Apoptosis , Caspasa 8/genética , Caspasa 8/inmunología , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata , Inflamación , Necrosis/inmunología , Necrosis/patología , Proteínas Quinasas/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
19.
Immunity ; 54(2): 247-258.e7, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33444549

RESUMEN

The vaccine strain against smallpox, vaccinia virus (VACV), is highly immunogenic yet causes relatively benign disease. These attributes are believed to be caused by gene loss in VACV. Using a targeted small interfering RNA (siRNA) screen, we identified a viral inhibitor found in cowpox virus (CPXV) and other orthopoxviruses that bound to the host SKP1-Cullin1-F-box (SCF) machinery and the essential necroptosis kinase receptor interacting protein kinase 3 (RIPK3). This "viral inducer of RIPK3 degradation" (vIRD) triggered ubiquitination and proteasome-mediated degradation of RIPK3 and inhibited necroptosis. In contrast to orthopoxviruses, the distantly related leporipoxvirus myxoma virus (MYXV), which infects RIPK3-deficient hosts, lacks a functional vIRD. Introduction of vIRD into VACV, which encodes a truncated and defective vIRD, enhanced viral replication in mice. Deletion of vIRD reduced CPXV-induced inflammation, viral replication, and mortality, which were reversed in RIPK3- and MLKL-deficient mice. Hence, vIRD-RIPK3 drives pathogen-host evolution and regulates virus-induced inflammation and pathogenesis.


Asunto(s)
Virus de la Viruela Vacuna/fisiología , Viruela Vacuna/inmunología , ARN Interferente Pequeño/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Virus Vaccinia/metabolismo , Proteínas Virales/metabolismo , Animales , Evolución Molecular , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inflamación , Ratones , Ratones Noqueados , Necroptosis/genética , Orthopoxvirus , Filogenia , Proteínas Quinasas/genética , Proteolisis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Análisis de Secuencia de ARN , Proteínas Virales/genética , Replicación Viral
20.
Annu Rev Genet ; 55: 235-263, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34813352

RESUMEN

The receptor-interacting protein kinase 1 (RIPK1) is recognized as a master upstream regulator that controls cell survival and inflammatory signaling as well as multiple cell death pathways, including apoptosis and necroptosis. The activation of RIPK1 kinase is extensively modulated by ubiquitination and phosphorylation, which are mediated by multiple factors that also control the activation of the NF-κB pathway. We discuss current findings regarding the genetic modulation of RIPK1 that controls its activation and interaction with downstream mediators, such as caspase-8 and RIPK3, to promote apoptosis and necroptosis. We also address genetic autoinflammatory human conditions that involve abnormal activation of RIPK1. Leveraging these new genetic and mechanistic insights, we postulate how an improved understanding of RIPK1 biology may support the development of therapeutics that target RIPK1 for the treatment of human inflammatory and neurodegenerative diseases.


Asunto(s)
Necroptosis , Proteínas Quinasas , Apoptosis/genética , Humanos , Necroptosis/genética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda