Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.148
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 25(7): 1193-1206, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38834865

ABSTRACT

Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKß-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.


Subject(s)
Cell Movement , Dendritic Cells , Homeostasis , Lymph Nodes , Mice, Inbred C57BL , Receptors, CCR7 , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymph Nodes/immunology , Lymph Nodes/cytology , Receptors, CCR7/metabolism , Mice , Cell Movement/immunology , Cell Shape , NF-kappa B/metabolism , Mice, Knockout , Signal Transduction/immunology , I-kappa B Kinase/metabolism , Actin-Related Protein 2-3 Complex/metabolism
2.
Cell ; 184(17): 4447-4463.e20, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34363755

ABSTRACT

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.


Subject(s)
Inflammation/enzymology , Protein Serine-Threonine Kinases/deficiency , Tumor Necrosis Factor-alpha/pharmacology , A549 Cells , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Autoimmunity/drug effects , Brain/diagnostic imaging , Cell Death/drug effects , Cytokines/metabolism , Deubiquitinating Enzyme CYLD/metabolism , Female , HEK293 Cells , Homozygote , Humans , I-kappa B Kinase/metabolism , Immunophenotyping , Inflammation/pathology , Interferon Type I/metabolism , Interferon-gamma/metabolism , Loss of Function Mutation/genetics , Male , Pedigree , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Pattern Recognition/metabolism , Toll-Like Receptor 3/metabolism , Transcriptome/genetics , Vesiculovirus/drug effects , Vesiculovirus/physiology
3.
Immunity ; 57(5): 929-932, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38749393

ABSTRACT

The ubiquitin-binding endoribonuclease N4BP1 is a critical immunosuppressor, but the mechanism by which it acts to constrain TLR-induced inflammatory cytokine production has remained unclear. In this issue of Immunity, Gitlin et al. find that N4BP1 works in concert with the non-canonical IκB kinase (IKK) to limit activity of the IKK complex.


Subject(s)
I-kappa B Kinase , Humans , I-kappa B Kinase/metabolism , Animals , Endoribonucleases/metabolism , Signal Transduction/immunology , Cytokines/metabolism
4.
Immunity ; 57(5): 973-986.e7, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38697117

ABSTRACT

The ubiquitin-binding endoribonuclease N4BP1 potently suppresses cytokine production by Toll-like receptors (TLRs) that signal through the adaptor MyD88 but is inactivated via caspase-8-mediated cleavage downstream of death receptors, TLR3, or TLR4. Here, we examined the mechanism whereby N4BP1 limits inflammatory responses. In macrophages, deletion of N4BP1 prolonged activation of inflammatory gene transcription at late time points after TRIF-independent TLR activation. Optimal suppression of inflammatory cytokines by N4BP1 depended on its ability to bind polyubiquitin chains, as macrophages and mice-bearing inactivating mutations in a ubiquitin-binding motif in N4BP1 displayed increased TLR-induced cytokine production. Deletion of the noncanonical IκB kinases (ncIKKs), Tbk1 and Ikke, or their adaptor Tank phenocopied N4bp1 deficiency and enhanced macrophage responses to TLR1/2, TLR7, or TLR9 stimulation. Mechanistically, N4BP1 acted in concert with the ncIKKs to limit the duration of canonical IκB kinase (IKKα/ß) signaling. Thus, N4BP1 and the ncIKKs serve as an important checkpoint against over-exuberant innate immune responses.


Subject(s)
Endoribonucleases , I-kappa B Kinase , Inflammation , Macrophages , Mice, Knockout , Protein Serine-Threonine Kinases , Signal Transduction , Toll-Like Receptors , Animals , Mice , Inflammation/immunology , Inflammation/metabolism , Toll-Like Receptors/metabolism , Macrophages/immunology , Macrophages/metabolism , I-kappa B Kinase/metabolism , I-kappa B Kinase/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Endoribonucleases/metabolism , Endoribonucleases/genetics , Ubiquitin/metabolism , Cytokines/metabolism , Mice, Inbred C57BL , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics
5.
Cell ; 174(6): 1477-1491.e19, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30146158

ABSTRACT

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.


Subject(s)
Apoptosis , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adult , Aged , Aging , Animals , Apoptosis/drug effects , Axons/metabolism , Behavior, Animal , Brain/cytology , Brain/metabolism , Cells, Cultured , Humans , I-kappa B Kinase/metabolism , Mice , Mice, Knockout , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Spinal Cord/metabolism , Staurosporine/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
6.
Mol Cell ; 84(13): 2436-2454.e10, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38925114

ABSTRACT

Signal transduction proteins containing a pLxIS motif induce interferon (IFN) responses central to antiviral immunity. Apart from their established roles in activating the IFN regulator factor (IRF) transcription factors, the existence of additional pathways and functions associated with the pLxIS motif is unknown. Using a synthetic biology-based platform, we identified two orphan pLxIS-containing proteins that stimulate IFN responses independent of all known pattern-recognition receptor pathways. We further uncovered a diversity of pLxIS signaling mechanisms, where the pLxIS motif represents one component of a multi-motif signaling entity, which has variable functions in activating IRF3, the TRAF6 ubiquitin ligase, IκB kinases, mitogen-activated protein kinases, and metabolic activities. The most diverse pLxIS signaling mechanisms were associated with the highest antiviral activities in human cells. The flexibility of domains that regulate IFN signaling may explain their prevalence in nature.


Subject(s)
Interferon Regulatory Factor-3 , Interferons , Signal Transduction , TNF Receptor-Associated Factor 6 , Humans , Interferons/metabolism , HEK293 Cells , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , I-kappa B Kinase/metabolism , I-kappa B Kinase/genetics , Protein Domains , Animals , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Amino Acid Motifs , Mitogen-Activated Protein Kinases/metabolism
7.
Immunity ; 55(12): 2271-2284.e7, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36384135

ABSTRACT

The NLRP3 inflammasome plays a central role in antimicrobial defense as well as in the context of sterile inflammatory conditions. NLRP3 activity is governed by two independent signals: the first signal primes NLRP3, rendering it responsive to the second signal, which then triggers inflammasome formation. Our understanding of how NLRP3 priming contributes to inflammasome activation remains limited. Here, we show that IKKß, a kinase activated during priming, induces recruitment of NLRP3 to phosphatidylinositol-4-phosphate (PI4P), a phospholipid enriched on the trans-Golgi network. NEK7, a mitotic spindle kinase that had previously been thought to be indispensable for NLRP3 activation, was redundant for inflammasome formation when IKKß recruited NLRP3 to PI4P. Studying iPSC-derived human macrophages revealed that the IKKß-mediated NEK7-independent pathway constitutes the predominant NLRP3 priming mechanism in human myeloid cells. Our results suggest that PI4P binding represents a primed state into which NLRP3 is brought by IKKß activity.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , I-kappa B Kinase , Inflammasomes/metabolism , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , trans-Golgi Network/metabolism
8.
Cell ; 167(4): 1052-1066.e18, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27814504

ABSTRACT

It is widely believed that inflammation associated with obesity has an important role in the development of type 2 diabetes. IκB kinase beta (IKKß) is a crucial kinase that responds to inflammatory stimuli such as tumor necrosis factor α (TNF-α) by initiating a variety of intracellular signaling cascades and is considered to be a key element in the inflammation-mediated development of insulin resistance. We show here, contrary to expectation, that IKKß-mediated inflammation is a positive regulator of hepatic glucose homeostasis. IKKß phosphorylates the spliced form of X-Box Binding Protein 1 (XBP1s) and increases the activity of XBP1s. We have used three experimental approaches to enhance the IKKß activity in the liver of obese mice and observed increased XBP1s activity, reduced ER stress, and a significant improvement in insulin sensitivity and consequently in glucose homeostasis. Our results reveal a beneficial role of IKKß-mediated hepatic inflammation in glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Glucose/metabolism , I-kappa B Kinase/metabolism , X-Box Binding Protein 1/metabolism , Animals , Cell Line, Tumor , Homeostasis , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Phosphorylation , Protein Stability
9.
Mol Cell ; 83(17): 3188-3204.e7, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37683611

ABSTRACT

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.


Subject(s)
NF-kappa B , Signal Transduction , NF-kappa B/genetics , I-kappa B Kinase/genetics , Protein Serine-Threonine Kinases/genetics , Mitochondria/genetics
10.
Mol Cell ; 82(13): 2415-2426.e5, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35477005

ABSTRACT

The NF-κB essential modulator (NEMO) is a regulatory subunit of the IκB kinase (IKK) complex that phosphorylates the NF-κB inhibitors IκBs. NEMO mediates IKK activation by binding to polyubiquitin chains (polyUb). Here, we show that Lys63(K63)-linked or linear polyUb binding to NEMO robustly induced the formation of liquid-like droplets in which IKK was activated. This liquid phase separation of NEMO was driven by multivalent interactions between NEMO and polyUb. Both the NEMO ubiquitin-binding (NUB) domain and the zinc-finger (ZF) domain of NEMO mediated binding to polyUb and contributed to NEMO phase separation and IKK activation in cells. Moreover, NEMO mutations associated with human immunodeficiency impaired its phase separation. These results demonstrate that polyUb activates IKK and NF-κB signaling by promoting the phase separation of NEMO.


Subject(s)
NF-kappa B , Polyubiquitin , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Polyubiquitin/genetics , Signal Transduction , Ubiquitin/metabolism
11.
Nat Immunol ; 18(7): 733-743, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28481329

ABSTRACT

The transcription regulator YAP controls organ size by regulating cell growth, proliferation and apoptosis. However, whether YAP has a role in innate antiviral immunity is largely unknown. Here we found that YAP negatively regulated an antiviral immune response. YAP deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in vivo. YAP blocked dimerization of the transcription factor IRF3 and impeded translocation of IRF3 to the nucleus after viral infection. Notably, virus-activated kinase IKKɛ phosphorylated YAP at Ser403 and thereby triggered degradation of YAP in lysosomes and, consequently, relief of YAP-mediated inhibition of the cellular antiviral response. These findings not only establish YAP as a modulator of the activation of IRF3 but also identify a previously unknown regulatory mechanism independent of the kinases Hippo and LATS via which YAP is controlled by the innate immune pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Fibroblasts/immunology , I-kappa B Kinase/metabolism , Immunity, Innate/immunology , Lysosomes/metabolism , Macrophages/immunology , Phosphoproteins/immunology , Rhabdoviridae Infections/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CRISPR-Cas Systems , Cell Cycle Proteins , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Fluorescent Antibody Technique , Gene Editing , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Lung/immunology , Lung/pathology , Mice , Microscopy, Confocal , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdoviridae Infections/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Vesiculovirus , Viral Load , YAP-Signaling Proteins
12.
Immunity ; 50(2): 348-361.e4, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30737145

ABSTRACT

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.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Thymocytes/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation/drug effects , I-kappa B Kinase/genetics , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Thymocytes/cytology , Thymocytes/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
13.
Cell ; 152(1-2): 304-15, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332762

ABSTRACT

The IκB kinase complex (IKK) is a key regulator of immune responses, inflammation, cell survival, and tumorigenesis. The prosurvival function of IKK centers on activation of the transcription factor NF-κB, whose target gene products inhibit caspases and prevent prolonged JNK activation. Here, we report that inactivation of the BH3-only protein BAD by IKK independently of NF-κB activation suppresses TNFα-induced apoptosis. TNFα-treated Ikkß(-/-) mouse embryonic fibroblasts (MEFs) undergo apoptosis significantly faster than MEFs deficient in both RelA and cRel due to lack of inhibition of BAD by IKK. IKK phosphorylates BAD at serine-26 (Ser26) and primes it for inactivation. Elimination of Ser26 phosphorylation promotes BAD proapoptotic activity, thereby accelerating TNFα-induced apoptosis in cultured cells and increasing mortality in animals. Our results reveal that IKK inhibits TNFα-induced apoptosis through two distinct but cooperative mechanisms: activation of the survival factor NF-κB and inactivation of the proapoptotic BH3-only BAD protein.


Subject(s)
Apoptosis , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , bcl-Associated Death Protein/metabolism , Animals , Fibroblasts/cytology , I-kappa B Kinase/genetics , Mice , Mice, Knockout , Phosphorylation , Serine/metabolism , bcl-Associated Death Protein/chemistry , bcl-Associated Death Protein/genetics , bcl-X Protein/metabolism
14.
EMBO J ; 42(20): e115307, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37691515

ABSTRACT

Removal of polyploid cells is essential to preventing cancer and restricting tumor growth. A new study published in The EMBO Journal shows assembly of the NEMO-PIDDosome on extra centrioles. Activation of this protein complex leads to NF-κB activation that, in turn, induces NK cell-mediated cell clearance.


Subject(s)
NF-kappa B , Signal Transduction , Humans , Gene Expression Regulation , I-kappa B Kinase/metabolism , Killer Cells, Natural , NF-kappa B/metabolism , Polyploidy
15.
EMBO J ; 42(21): e114719, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37737566

ABSTRACT

Activation of the IκB kinase (IKK) complex has recurrently been linked to colorectal cancer (CRC) initiation and progression. However, identification of downstream effectors other than NF-κB has remained elusive. Here, analysis of IKK-dependent substrates in CRC cells after UV treatment revealed that phosphorylation of BRD4 by IKK-α is required for its chromatin-binding at target genes upon DNA damage. Moreover, IKK-α induces the NF-κB-dependent transcription of the cytokine LIF, leading to STAT3 activation, association with BRD4 and recruitment to specific target genes. IKK-α abrogation results in defective BRD4 and STAT3 functions and consequently irreparable DNA damage and apoptotic cell death upon different stimuli. Simultaneous inhibition of BRAF-dependent IKK-α activity, BRD4, and the JAK/STAT pathway enhanced the therapeutic potential of 5-fluorouracil combined with irinotecan in CRC cells and is curative in a chemotherapy-resistant xenograft model. Finally, coordinated expression of LIF and IKK-α is a poor prognosis marker for CRC patients. Our data uncover a functional link between IKK-α, BRD4, and JAK/STAT signaling with clinical relevance.


Subject(s)
I-kappa B Kinase , Signal Transduction , Humans , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Janus Kinases/genetics , STAT Transcription Factors , Phosphorylation , Tumor Necrosis Factor-alpha/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
16.
Nat Immunol ; 16(12): 1235-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502405

ABSTRACT

Ectopic lymphoid-like structures (ELSs) are often observed in cancer, yet their function is obscure. Although ELSs signify good prognosis in certain malignancies, we found that hepatic ELSs indicated poor prognosis for hepatocellular carcinoma (HCC). We studied an HCC mouse model that displayed abundant ELSs and found that they constituted immunopathological microniches wherein malignant hepatocyte progenitor cells appeared and thrived in a complex cellular and cytokine milieu until gaining self-sufficiency. The egress of progenitor cells and tumor formation were associated with the autocrine production of cytokines previously provided by the niche. ELSs developed via cooperation between the innate immune system and adaptive immune system, an event facilitated by activation of the transcription factor NF-κB and abolished by depletion of T cells. Such aberrant immunological foci might represent new targets for cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/immunology , Liver Neoplasms/immunology , Lymphoid Tissue/immunology , Neoplastic Stem Cells/immunology , Stem Cell Niche/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Comparative Genomic Hybridization , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunoblotting , In Situ Hybridization , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Niche/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
17.
Immunity ; 49(2): 195-197, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30134194

ABSTRACT

Insect immunity to extracellular microbes relies largely on the TOLL and IMD pathways. In this issue of Immunity, Goto et al. (2018) report that the IKKß-Relish module of the IMD pathway hitches up the intracellular sensor STING to activate antiviral responses in Drosophila.


Subject(s)
Antiviral Agents , Drosophila Proteins , Animals , Drosophila , I-kappa B Kinase , NF-kappa B , Signal Transduction
18.
Immunity ; 49(2): 225-234.e4, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30119996

ABSTRACT

Antiviral immunity in Drosophila involves RNA interference and poorly characterized inducible responses. Here, we showed that two components of the IMD pathway, the kinase dIKKß and the transcription factor Relish, were required to control infection by two picorna-like viruses. We identified a set of genes induced by viral infection and regulated by dIKKß and Relish, which included an ortholog of STING. We showed that dSTING participated in the control of infection by picorna-like viruses, acting upstream of dIKKß to regulate expression of Nazo, an antiviral factor. Our data reveal an antiviral function for STING in an animal model devoid of interferons and suggest an evolutionarily ancient role for this molecule in antiviral immunity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , I-kappa B Kinase/metabolism , Membrane Proteins/metabolism , Peptide Initiation Factors/metabolism , Picornaviridae Infections/immunology , Animals , Cell Line , Dicistroviridae/immunology , Drosophila Proteins/genetics , I-kappa B Kinase/genetics , Membrane Proteins/genetics , Peptide Initiation Factors/genetics , RNA Interference , Transcription Factors/metabolism
19.
Mol Cell ; 76(1): 148-162.e7, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31447391

ABSTRACT

The rapid proliferation of cancer cells and dysregulated vasculature within the tumor leads to limited nutrient accessibility. Cancer cells often rewire their metabolic pathways for adaption to nutrient stress, and the underlying mechanism remains largely unknown. Glutamate dehydrogenase 1 (GDH1) is a key enzyme in glutaminolysis that converts glutamate to α-ketoglutarate (α-KG). Here, we show that, under low glucose, GDH1 is phosphorylated at serine (S) 384 and interacts with RelA and IKKß. GDH1-produced α-KG directly binds to and activates IKKß and nuclear factor κB (NF-κB) signaling, which promotes glucose uptake and tumor cell survival by upregulating GLUT1, thereby accelerating gliomagenesis. In addition, GDH1 S384 phosphorylation correlates with the malignancy and prognosis of human glioblastoma. Our finding reveals a unique role of α-KG to directly regulate signal pathway, uncovers a distinct mechanism of metabolite-mediated NF-κB activation, and also establishes the critical role of α-KG-activated NF-κB in brain tumor development.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation , Energy Metabolism , Glioblastoma/metabolism , Glucose/metabolism , Glutamate Dehydrogenase/metabolism , Ketoglutaric Acids/metabolism , NF-kappa B/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Child , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Glucose/deficiency , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glutamate Dehydrogenase/genetics , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NF-kappa B/genetics , Neoplasm Grading , Phosphorylation , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Young Adult
20.
Mol Cell ; 75(4): 669-682.e5, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31302002

ABSTRACT

Phosphorylated IKKα(p45) is a nuclear active form of the IKKα kinase that is induced by the MAP kinases BRAF and TAK1 and promotes tumor growth independent of canonical NF-κB signaling. Insights into the sources of IKKα(p45) activation and its downstream substrates in the nucleus remain to be defined. Here, we discover that IKKα(p45) is rapidly activated by DNA damage independent of ATM-ATR, but dependent on BRAF-TAK1-p38-MAPK, and is required for robust ATM activation and efficient DNA repair. Abolishing BRAF or IKKα activity attenuates ATM, Chk1, MDC1, Kap1, and 53BP1 phosphorylation, compromises 53BP1 and RIF1 co-recruitment to sites of DNA lesions, and inhibits 53BP1-dependent fusion of dysfunctional telomeres. Furthermore, IKKα or BRAF inhibition synergistically enhances the therapeutic potential of 5-FU and irinotecan to eradicate chemotherapy-resistant metastatic human tumors in vivo. Our results implicate BRAF and IKKα kinases in the DDR and reveal a combination strategy for cancer treatment.


Subject(s)
DNA Damage , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , I-kappa B Kinase/metabolism , Irinotecan/pharmacology , MAP Kinase Signaling System , Neoplasm Proteins , Neoplasms , Animals , DNA Repair/drug effects , DNA Repair/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , HCT116 Cells , Humans , I-kappa B Kinase/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Telomere/genetics , Telomere/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL