Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.522
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 179(3): 787-799.e17, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31626775

RESUMEN

Genetic screens are critical for the systematic identification of genes underlying cellular phenotypes. Pooling gene perturbations greatly improves scalability but is not compatible with imaging of complex and dynamic cellular phenotypes. Here, we introduce a pooled approach for optical genetic screens in mammalian cells. We use targeted in situ sequencing to demultiplex a library of genetic perturbations following image-based phenotyping. We screened a set of 952 genes across millions of cells for involvement in nuclear factor κB (NF-κB) signaling by imaging the translocation of RelA (p65) to the nucleus. Screening at a single time point across 3 cell lines recovered 15 known pathway components, while repeating the screen with live-cell imaging revealed a role for Mediator complex subunits in regulating the duration of p65 nuclear retention. These results establish a highly multiplexed approach to image-based screens of spatially and temporally defined phenotypes with pooled libraries.


Asunto(s)
Pruebas Genéticas , Genómica , FN-kappa B/genética , Factor de Transcripción ReIA/genética , Animales , Sistemas CRISPR-Cas , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Humanos , Complejo Mediador/genética , ARN Guía de Kinetoplastida/genética
2.
Cell ; 172(4): 731-743.e12, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29425491

RESUMEN

The noncanonical IKK family member TANK-binding kinase 1 (TBK1) is activated by pro-inflammatory cytokines, but its role in controlling metabolism remains unclear. Here, we report that the kinase uniquely controls energy metabolism. Tbk1 expression is increased in adipocytes of HFD-fed mice. Adipocyte-specific TBK1 knockout (ATKO) attenuates HFD-induced obesity by increasing energy expenditure; further studies show that TBK1 directly inhibits AMPK to repress respiration and increase energy storage. Conversely, activation of AMPK under catabolic conditions can increase TBK1 activity through phosphorylation, mediated by AMPK's downstream target ULK1. Surprisingly, ATKO also exaggerates adipose tissue inflammation and insulin resistance. TBK1 suppresses inflammation by phosphorylating and inducing the degradation of the IKK kinase NIK, thus attenuating NF-κB activity. Moreover, TBK1 mediates the negative impact of AMPK activity on NF-κB activation. These data implicate a unique role for TBK1 in mediating bidirectional crosstalk between energy sensing and inflammatory signaling pathways in both over- and undernutrition.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Metabolismo Energético , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/patología , Tejido Adiposo/patología , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular Transformada , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Serina-Treonina Quinasas/genética , Quinasa de Factor Nuclear kappa B
3.
Annu Rev Biochem ; 86: 715-748, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28441057

RESUMEN

Oxidative stress is two sided: Whereas excessive oxidant challenge causes damage to biomolecules, maintenance of a physiological level of oxidant challenge, termed oxidative eustress, is essential for governing life processes through redox signaling. Recent interest has focused on the intricate ways by which redox signaling integrates these converse properties. Redox balance is maintained by prevention, interception, and repair, and concomitantly the regulatory potential of molecular thiol-driven master switches such as Nrf2/Keap1 or NF-κB/IκB is used for system-wide oxidative stress response. Nonradical species such as hydrogen peroxide (H2O2) or singlet molecular oxygen, rather than free-radical species, perform major second messenger functions. Chemokine-controlled NADPH oxidases and metabolically controlled mitochondrial sources of H2O2 as well as glutathione- and thioredoxin-related pathways, with powerful enzymatic back-up systems, are responsible for fine-tuning physiological redox signaling. This makes for a rich research field spanning from biochemistry and cell biology into nutritional sciences, environmental medicine, and molecular knowledge-based redox medicine.


Asunto(s)
Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Regulación de la Expresión Génica , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , NADPH Oxidasas/genética , Factor 2 Relacionado con NF-E2/genética , Inhibidor NF-kappaB alfa/genética , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/genética , Oxidación-Reducción , Transducción de Señal , Oxígeno Singlete/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
4.
Nat Immunol ; 21(5): 535-545, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32313245

RESUMEN

Despite evidence of chronic inflammation in myelodysplastic syndrome (MDS) and cell-intrinsic dysregulation of Toll-like receptor (TLR) signaling in MDS hematopoietic stem and progenitor cells (HSPCs), the mechanisms responsible for the competitive advantage of MDS HSPCs in an inflammatory milieu over normal HSPCs remain poorly defined. Here, we found that chronic inflammation was a determinant for the competitive advantage of MDS HSPCs and for disease progression. The cell-intrinsic response of MDS HSPCs, which involves signaling through the noncanonical NF-κB pathway, protected these cells from chronic inflammation as compared to normal HSPCs. In response to inflammation, MDS HSPCs switched from canonical to noncanonical NF-κB signaling, a process that was dependent on TLR-TRAF6-mediated activation of A20. The competitive advantage of TLR-TRAF6-primed HSPCs could be restored by deletion of A20 or inhibition of the noncanonical NF-κB pathway. These findings uncover the mechanistic basis for the clonal dominance of MDS HSPCs and indicate that interfering with noncanonical NF-κB signaling could prevent MDS progression.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Inflamación/inmunología , Síndromes Mielodisplásicos/inmunología , FN-kappa B/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Anciano , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Transgénicos , Mielopoyesis , FN-kappa B/genética , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/genética , Receptores Toll-Like/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
5.
Cell ; 168(1-2): 37-57, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28086098

RESUMEN

NF-κB was discovered 30 years ago as a rapidly inducible transcription factor. Since that time, it has been found to have a broad role in gene induction in diverse cellular responses, particularly throughout the immune system. Here, we summarize elaborate regulatory pathways involving this transcription factor and use recent discoveries in human genetic diseases to place specific proteins within their relevant medical and biological contexts.


Asunto(s)
Inflamación/metabolismo , FN-kappa B/historia , FN-kappa B/metabolismo , Animales , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Inflamación/inmunología , Ratones , Mutación , FN-kappa B/química , FN-kappa B/genética , Transducción de Señal
6.
Mol Cell ; 84(6): 1149-1157.e7, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38309274

RESUMEN

OCA-B, OCA-T1, and OCA-T2 belong to a family of coactivators that bind to POU transcription factors (TFs) to regulate gene expression in immune cells. Here, we identify IκBζ (encoded by the NFKBIZ gene) as an additional coactivator of POU TFs. Although originally discovered as an inducible regulator of NF-κB, we show here that IκBζ shares a microhomology with OCA proteins and uses this segment to bind to POU TFs and octamer-motif-containing DNA. Our functional experiments suggest that IκBζ requires its interaction with POU TFs to coactivate immune-related genes. This finding is reinforced by epigenomic analysis of MYD88L265P-mutant lymphoma cells, which revealed colocalization of IκBζ with the POU TF OCT2 and NF-κB:p50 at hundreds of DNA elements harboring octamer and κB motifs. These results suggest that IκBζ is a transcriptional coactivator that can amplify and integrate the output of NF-κB and POU TFs at inducible genes in immune cells.


Asunto(s)
ADN , FN-kappa B , FN-kappa B/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , ADN/genética , ADN/metabolismo
7.
Mol Cell ; 84(3): 522-537.e8, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38151017

RESUMEN

The anti-cancer target hRpn13 is a proteasome substrate receptor. However, hRpn13-targeting molecules do not impair its interaction with proteasomes or ubiquitin, suggesting other critical cellular activities. We find that hRpn13 depletion causes correlated proteomic and transcriptomic changes, with pronounced effects in myeloma cells for cytoskeletal and immune response proteins and bone-marrow-specific arginine deiminase PADI4. Moreover, a PROTAC against hRpn13 co-depletes PADI4, histone deacetylase HDAC8, and DNA methyltransferase MGMT. PADI4 binds and citrullinates hRpn13 and proteasomes, and proteasomes from PADI4-inhibited myeloma cells exhibit reduced peptidase activity. When off proteasomes, hRpn13 can bind HDAC8, and this interaction inhibits HDAC8 activity. Further linking hRpn13 to transcription, its loss reduces nuclear factor κB (NF-κB) transcription factor p50, which proteasomes generate by cleaving its precursor protein. NF-κB inhibition depletes hRpn13 interactors PADI4 and HDAC8. Altogether, we find that hRpn13 acts dually in protein degradation and expression and that proteasome constituency and, in turn, regulation varies by cell type.


Asunto(s)
Histona Desacetilasas , Péptidos y Proteínas de Señalización Intracelular , FN-kappa B , Arginina Deiminasa Proteína-Tipo 4 , Factores de Transcripción , Humanos , Epigénesis Genética , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteoma/metabolismo , Proteómica , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Línea Celular Tumoral
8.
Mol Cell ; 84(17): 3271-3287.e8, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39178863

RESUMEN

Cellular senescence, a stress-induced stable proliferation arrest associated with an inflammatory senescence-associated secretory phenotype (SASP), is a cause of aging. In senescent cells, cytoplasmic chromatin fragments (CCFs) activate SASP via the anti-viral cGAS/STING pathway. Promyelocytic leukemia (PML) protein organizes PML nuclear bodies (NBs), which are also involved in senescence and anti-viral immunity. The HIRA histone H3.3 chaperone localizes to PML NBs in senescent cells. Here, we show that HIRA and PML are essential for SASP expression, tightly linked to HIRA's localization to PML NBs. Inactivation of HIRA does not directly block expression of nuclear factor κB (NF-κB) target genes. Instead, an H3.3-independent HIRA function activates SASP through a CCF-cGAS-STING-TBK1-NF-κB pathway. HIRA physically interacts with p62/SQSTM1, an autophagy regulator and negative SASP regulator. HIRA and p62 co-localize in PML NBs, linked to their antagonistic regulation of SASP, with PML NBs controlling their spatial configuration. These results outline a role for HIRA and PML in the regulation of SASP.


Asunto(s)
Proteínas de Ciclo Celular , Senescencia Celular , Chaperonas de Histonas , Inflamación , FN-kappa B , Proteínas Nucleares , Proteína de la Leucemia Promielocítica , Proteínas Serina-Treonina Quinasas , Proteína Sequestosoma-1 , Transducción de Señal , Factores de Transcripción , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Autofagia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Cromatina/metabolismo , Cromatina/genética , Células HEK293 , Chaperonas de Histonas/metabolismo , Chaperonas de Histonas/genética , Histonas/metabolismo , Histonas/genética , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , FN-kappa B/metabolismo , FN-kappa B/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Nucleotidiltransferasas , Proteína de la Leucemia Promielocítica/metabolismo , Proteína de la Leucemia Promielocítica/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
9.
Mol Cell ; 83(17): 3188-3204.e7, 2023 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-37683611

RESUMEN

Failure to clear damaged mitochondria via mitophagy disrupts physiological function and may initiate damage signaling via inflammatory cascades, although how these pathways intersect remains unclear. We discovered that nuclear factor kappa B (NF-κB) essential regulator NF-κB effector molecule (NEMO) is recruited to damaged mitochondria in a Parkin-dependent manner in a time course similar to recruitment of the structurally related mitophagy adaptor, optineurin (OPTN). Upon recruitment, NEMO partitions into phase-separated condensates distinct from OPTN but colocalizing with p62/SQSTM1. NEMO recruitment, in turn, recruits the active catalytic inhibitor of kappa B kinase (IKK) component phospho-IKKß, initiating NF-κB signaling and the upregulation of inflammatory cytokines. Consistent with a potential neuroinflammatory role, NEMO is recruited to mitochondria in primary astrocytes upon oxidative stress. These findings suggest that damaged, ubiquitinated mitochondria serve as an intracellular platform to initiate innate immune signaling, promoting the formation of activated IKK complexes sufficient to activate NF-κB signaling. We propose that mitophagy and NF-κB signaling are initiated as parallel pathways in response to mitochondrial stress.


Asunto(s)
FN-kappa B , Transducción de Señal , FN-kappa B/genética , Quinasa I-kappa B/genética , Proteínas Serina-Treonina Quinasas/genética , Mitocondrias/genética
10.
Immunity ; 54(5): 916-930.e7, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979588

RESUMEN

Macrophages initiate inflammatory responses via the transcription factor NFκB. The temporal pattern of NFκB activity determines which genes are expressed and thus, the type of response that ensues. Here, we examined how information about the stimulus is encoded in the dynamics of NFκB activity. We generated an mVenus-RelA reporter mouse line to enable high-throughput live-cell analysis of primary macrophages responding to host- and pathogen-derived stimuli. An information-theoretic workflow identified six dynamical features-termed signaling codons-that convey stimulus information to the nucleus. In particular, oscillatory trajectories were a hallmark of responses to cytokine but not pathogen-derived stimuli. Single-cell imaging and RNA sequencing of macrophages from a mouse model of Sjögren's syndrome revealed inappropriate responses to stimuli, suggestive of confusion of two NFκB signaling codons. Thus, the dynamics of NFκB signaling classify immune threats through six signaling codons, and signal confusion based on defective codon deployment may underlie the etiology of some inflammatory diseases.


Asunto(s)
Codón/genética , Macrófagos/fisiología , FN-kappa B/genética , Transducción de Señal/genética , Animales , Células Cultivadas , Citocinas/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Síndrome de Sjögren/genética , Factor de Transcripción ReIA/genética
11.
Nature ; 623(7988): 803-813, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37938781

RESUMEN

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Asunto(s)
Autoanticuerpos , Predisposición Genética a la Enfermedad , Interferón Tipo I , FN-kappa B , Humanos , Autoanticuerpos/inmunología , COVID-19/genética , COVID-19/inmunología , Mutación con Ganancia de Función , Heterocigoto , Proteínas I-kappa B/deficiencia , Proteínas I-kappa B/genética , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Mutación con Pérdida de Función , FN-kappa B/deficiencia , FN-kappa B/genética , Subunidad p52 de NF-kappa B/deficiencia , Subunidad p52 de NF-kappa B/genética , Neumonía Viral/genética , Neumonía Viral/inmunología , Timo/anomalías , Timo/inmunología , Timo/patología , Células Epiteliales Tiroideas/metabolismo , Células Epiteliales Tiroideas/patología , Proteína AIRE , Quinasa de Factor Nuclear kappa B
12.
EMBO J ; 43(5): 780-805, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38316991

RESUMEN

Inflammation is a common condition of prostate tissue, whose impact on carcinogenesis is highly debated. Microbial colonization is a well-documented cause of a small percentage of prostatitis cases, but it remains unclear what underlies the majority of sterile inflammation reported. Here, androgen- independent fluctuations of PSA expression in prostate cells have lead us to identify a prominent function of the Transient Receptor Potential Cation Channel Subfamily M Member 8 (TRPM8) gene in sterile inflammation. Prostate cells secret TRPM8 RNA into extracellular vesicles (EVs), which primes TLR3/NF-kB-mediated inflammatory signaling after EV endocytosis by epithelial cancer cells. Furthermore, prostate cancer xenografts expressing a translation-defective form of TRPM8 RNA contain less collagen type I in the extracellular matrix, significantly more infiltrating NK cells, and larger necrotic areas as compared to control xenografts. These findings imply sustained, androgen-independent expression of TRPM8 constitutes as a promoter of anticancer innate immunity, which may constitute a clinically relevant condition affecting prostate cancer prognosis.


Asunto(s)
Neoplasias de la Próstata , Canales Catiónicos TRPM , Humanos , Masculino , Andrógenos , Inflamación/genética , Factor 3 Regulador del Interferón , Proteínas de la Membrana , FN-kappa B/genética , Neoplasias de la Próstata/genética , Receptor Toll-Like 3/genética , Canales Catiónicos TRPM/genética , Animales
13.
EMBO J ; 43(6): 904-930, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38337057

RESUMEN

Mitochondrial outer membrane permeabilisation (MOMP) is often essential for apoptosis, by enabling cytochrome c release that leads to caspase activation and rapid cell death. Recently, MOMP has been shown to be inherently pro-inflammatory with emerging cellular roles, including its ability to elicit anti-tumour immunity. Nonetheless, how MOMP triggers inflammation and how the cell regulates this remains poorly defined. We find that upon MOMP, many proteins localised either to inner or outer mitochondrial membranes are ubiquitylated in a promiscuous manner. This extensive ubiquitylation serves to recruit the essential adaptor molecule NEMO, leading to the activation of pro-inflammatory NF-κB signalling. We show that disruption of mitochondrial outer membrane integrity through different means leads to the engagement of a similar pro-inflammatory signalling platform. Therefore, mitochondrial integrity directly controls inflammation, such that permeabilised mitochondria initiate NF-κB signalling.


Asunto(s)
FN-kappa B , Ubiquitina , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Ubiquitina/metabolismo , Membranas Mitocondriales/metabolismo , Mitocondrias/metabolismo , Apoptosis/fisiología , Inflamación/metabolismo
14.
EMBO J ; 43(5): 806-835, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38287188

RESUMEN

In mammalian somatic cells, the relative contribution of RNAi and the type I interferon response during viral infection is unclear. The apparent inefficiency of antiviral RNAi might be due to self-limiting properties and mitigating co-factors of the key enzyme Dicer. In particular, the helicase domain of human Dicer appears to be an important restriction factor of its activity. Here, we study the involvement of several helicase-truncated mutants of human Dicer in the antiviral response. All deletion mutants display a PKR-dependent antiviral phenotype against certain viruses, and one of them, Dicer N1, acts in a completely RNAi-independent manner. Transcriptomic analyses show that many genes from the interferon and inflammatory response pathways are upregulated in Dicer N1 expressing cells. We show that some of these genes are controlled by NF-kB and that blocking this pathway abrogates the antiviral phenotype of Dicer N1. Our findings highlight the crosstalk between Dicer, PKR, and the NF-kB pathway, and suggest that human Dicer may have repurposed its helicase domain to prevent basal activation of antiviral and inflammatory pathways.


Asunto(s)
ARN Helicasas DEAD-box , Interferón Tipo I , FN-kappa B , Infecciones por Virus ARN , Ribonucleasa III , Animales , Humanos , FN-kappa B/genética , Interferencia de ARN , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Ribonucleasa III/química , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Infecciones por Virus ARN/enzimología
15.
Nat Immunol ; 17(10): 1216-1225, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27500631

RESUMEN

The epigenetic processes that regulate antibody-secreting plasma cells are not well understood. Here, analysis of plasma cell differentiation revealed DNA hypomethylation of 10% of CpG loci that were overrepresented at enhancers. Inhibition of DNA methylation enhanced plasma cell commitment in a cell-division-dependent manner. Analysis of B cells differentiating in vivo stratified by cell division revealed a fivefold increase in mRNA transcription coupled to DNA hypomethylation. Demethylation occurred first at binding motifs for the transcription factors NF-κB and AP-1 and later at those for the transcription factors IRF and Oct-2 and was coincident with activation and differentiation gene-expression programs in a cell-division-dependent manner. These data provide mechanistic insight into cell-division-coupled transcriptional and epigenetic reprogramming and suggest that DNA hypomethylation reflects the cis-regulatory history of plasma cell differentiation.


Asunto(s)
Linfocitos B/fisiología , Metilación de ADN , FN-kappa B/metabolismo , Células Plasmáticas/fisiología , Factor de Transcripción AP-1/metabolismo , Animales , Sitios de Unión/genética , Diferenciación Celular/genética , División Celular/genética , Células Cultivadas , Islas de CpG/genética , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Inmunidad Humoral/genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Factor 2 de Transcripción de Unión a Octámeros/genética , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Factor de Transcripción AP-1/genética
16.
Nat Immunol ; 17(5): 565-73, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27043411

RESUMEN

Positive selection occurs in the thymic cortex, but critical maturation events occur later in the medulla. Here we defined the precise stage at which T cells acquired competence to proliferate and emigrate. Transcriptome analysis of late gene changes suggested roles for the transcription factor NF-κB and interferon signaling. Mice lacking the inhibitor of NF-κB (IκB) kinase (IKK) kinase TAK1 underwent normal positive selection but exhibited a specific block in functional maturation. NF-κB signaling provided protection from death mediated by the cytokine TNF and was required for proliferation and emigration. The interferon signature was independent of NF-κB; however, thymocytes deficient in the interferon-α (IFN-α) receptor IFN-αR showed reduced expression of the transcription factor STAT1 and phenotypic abnormality but were able to proliferate. Thus, both NF-κB and tonic interferon signals are involved in the final maturation of thymocytes into naive T cells.


Asunto(s)
Diferenciación Celular/inmunología , FN-kappa B/inmunología , Receptor de Interferón alfa y beta/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Movimiento Celular/inmunología , Proliferación Celular/genética , Citometría de Flujo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptor de Interferón alfa y beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Linfocitos T/metabolismo , Timocitos/inmunología , Timocitos/metabolismo , Timo/citología , Timo/metabolismo , Transcriptoma/genética , Transcriptoma/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
17.
Immunity ; 50(2): 348-361.e4, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30737145

RESUMEN

NF-κB (nuclear factor κB) signaling is considered critical for single positive (SP) thymocyte development because loss of upstream activators of NF-κB, such as the IKK complex, arrests their development. We found that the compound ablation of RelA, cRel, and p50, required for canonical NF-κB transcription, had no impact upon thymocyte development. While IKK-deficient thymocytes were acutely sensitive to tumor necrosis factor (TNF)-induced cell death, Rel-deficient cells remained resistant, calling into question the importance of NF-κB as the IKK target required for thymocyte survival. Instead, we found that IKK controlled thymocyte survival by repressing cell-death-inducing activity of the serine/threonine kinase RIPK1. We observed that RIPK1 expression was induced during development of SP thymocytes and that IKK was required to prevent RIPK1-kinase-dependent death of SPs in vivo. Finally, we showed that IKK was required to protect Rel-deficient thymocytes from RIPK1-dependent cell death, underscoring the NF-κB-independent function of IKK during thymic development.


Asunto(s)
Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Timocitos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Quinasa I-kappa B/genética , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Timocitos/citología , Timocitos/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
18.
Mol Cell ; 80(4): 555-557, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33217313

RESUMEN

In this issue of Molecular Cell, Byun et al. (2020) find that the dual targeting of glutamine metabolism and the PD-L1 checkpoint inhibitor augments anti-tumor immunity. Mechanistically, decreased glutamine availability attenuated S-glutathionylation of SERCA, resulting in an increase in cytosolic calcium, enhanced NF-κB activity, and upregulation of programmed death-ligand 1.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Antígeno B7-H1/genética , Glutamina , Humanos , FN-kappa B/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética
19.
EMBO J ; 42(15): e113126, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37345898

RESUMEN

N6 -methyladenosine (m6 A) in messenger RNA (mRNA) regulates immune cells in homeostasis and in response to infection and inflammation. The function of the m6 A reader YTHDF2 in the tumor microenvironment (TME) in these contexts has not been explored. We discovered that the loss of YTHDF2 in regulatory T (Treg) cells reduces tumor growth in mice. Deletion of Ythdf2 in Tregs does not affect peripheral immune homeostasis but leads to increased apoptosis and impaired suppressive function of Treg cells in the TME. Elevated tumor necrosis factor (TNF) signaling in the TME promotes YTHDF2 expression, which in turn regulates NF-κB signaling by accelerating the degradation of m6 A-modified transcripts that encode NF-κB-negative regulators. This TME-specific regulation of Treg by YTHDF2 points to YTHDF2 as a potential target for anti-cancer immunotherapy, where intratumoral Treg cells can be targeted to enhance anti-tumor immune response while avoiding Treg cells in the periphery to minimize undesired inflammations.


Asunto(s)
FN-kappa B , Neoplasias , Ratones , Animales , FN-kappa B/genética , Neoplasias/genética , Transducción de Señal , Inmunoterapia , Inflamación , Microambiente Tumoral
20.
EMBO J ; 42(20): e113510, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37530438

RESUMEN

Unscheduled increases in ploidy underlie defects in tissue function, premature aging, and malignancy. A concomitant event to polyploidization is the amplification of centrosomes, the main microtubule organization centers in animal cells. Supernumerary centrosomes are frequent in tumors, correlating with higher aggressiveness and poor prognosis. However, extra centrosomes initially also exert an onco-protective effect by activating p53-induced cell cycle arrest. If additional signaling events initiated by centrosomes help prevent pathology is unknown. Here, we report that extra centrosomes, arising during unscheduled polyploidization or aberrant centriole biogenesis, induce activation of NF-κB signaling and sterile inflammation. This signaling requires the NEMO-PIDDosome, a multi-protein complex composed of PIDD1, RIPK1, and NEMO/IKKγ. Remarkably, the presence of supernumerary centrosomes suffices to induce a paracrine chemokine and cytokine profile, able to polarize macrophages into a pro-inflammatory phenotype. Furthermore, extra centrosomes increase the immunogenicity of cancer cells and render them more susceptible to NK-cell attack. Hence, the PIDDosome acts as a dual effector, able to engage not only the p53 network for cell cycle control but also NF-κB signaling to instruct innate immunity.


Asunto(s)
FN-kappa B , Neoplasias , Animales , Centrosoma/metabolismo , Inflamación/patología , Monitorización Inmunológica , Neoplasias/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA