Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Sci Adv ; 10(3): eadh5520, 2024 Jan 19.
Article En | MEDLINE | ID: mdl-38232171

Acute thymic atrophy occurs following type 1 inflammatory conditions such as viral infection and sepsis, resulting in cell death and disruption of T cell development. However, the impact type 1 immunity has on thymic-resident innate lymphoid cells (ILCs) remains unclear. Single-cell RNA sequencing revealed neonatal thymic-resident type 1 ILCs (ILC1s) as a unique and immature subset compared to ILC1s in other primary lymphoid organs. Culturing murine neonatal thymic lobes with the type 1 cytokines interleukin-12 (IL-12) and IL-18 resulted in a rapid expansion and thymic egress of KLRG1+CXCR6+ cytotoxic ILC1s. Live imaging showed the subcapsular thymic localization and exit of ILC1s following IL-12 + IL-18 stimulation. Similarly, murine cytomegalovirus infection in neonates resulted in thymic atrophy and subcapsular localization of thymic-resident ILC1s. Neonatal thymic grafting revealed that type 1 inflammation enhances the homing of cytokine-producing thymus-derived ILC1s to the liver and peritoneal cavity. Together, we show that type 1 immunity promotes the expansion and peripheral homing of thymic-derived ILC1s.


Interleukin-18 , Lymphocytes , Humans , Infant, Newborn , Mice , Animals , Lymphocytes/metabolism , Immunity, Innate , Cytokines/metabolism , Interleukin-12 , Atrophy
2.
Sci Immunol ; 8(83): eadd3955, 2023 05 12.
Article En | MEDLINE | ID: mdl-37172103

Dendritic cells (DCs) mature in an immunogenic or tolerogenic manner depending on the context in which an antigen is perceived, preserving the balance between immunity and tolerance. Whereas the pathways driving immunogenic maturation in response to infectious insults are well-characterized, the signals that drive tolerogenic maturation during homeostasis are still poorly understood. We found that the engulfment of apoptotic cells triggered homeostatic maturation of type 1 conventional DCs (cDC1s) within the spleen. This maturation process could be mimicked by engulfment of empty, nonadjuvanted lipid nanoparticles (LNPs), was marked by intracellular accumulation of cholesterol, and was highly specific to cDC1s. Engulfment of either apoptotic cells or cholesterol-rich LNPs led to the activation of the liver X receptor (LXR) pathway, which promotes the efflux of cellular cholesterol, and repressed genes associated with immunogenic maturation. In contrast, simultaneous engagement of TLR3 to mimic viral infection via administration of poly(I:C)-adjuvanted LNPs repressed the LXR pathway, thus delaying cellular cholesterol efflux and inducing genes that promote T cell-mediated immunity. These data demonstrate that conserved cellular cholesterol efflux pathways are differentially regulated in tolerogenic versus immunogenic cDC1s and suggest that administration of nonadjuvanted cholesterol-rich LNPs may be an approach for inducing tolerogenic DC maturation.


Dendritic Cells , Signal Transduction , Liver X Receptors/metabolism , Signal Transduction/genetics , Homeostasis , Cholesterol
3.
iScience ; 26(12): 108570, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-38162021

The unfolded protein response (UPR) aims to restore ER homeostasis under conditions of high protein folding load, a function primarily serving secretory cells. Additional, non-canonical UPR functions have recently been unraveled in immune cells. We addressed the function of the inositol-requiring enzyme 1 (IRE1) signaling branch of the UPR in NK cells in homeostasis and microbial challenge. Cell-intrinsic compound deficiency of IRE1 and its downstream transcription factor XBP1 in NKp46+ NK cells, did not affect basal NK cell homeostasis, or overall outcome of viral MCMV infection. However, mixed bone marrow chimeras revealed a competitive advantage in the proliferation of IRE1-sufficient Ly49H+ NK cells after viral infection. CITE-Seq analysis confirmed strong induction of IRE1 early upon infection, concomitant with the activation of a canonical UPR signature. Therefore, we conclude that IRE1/XBP1 activation is required during vigorous NK cell proliferation early upon viral infection, as part of a canonical UPR response.

4.
Nat Immunol ; 21(4): 381-387, 2020 04.
Article En | MEDLINE | ID: mdl-32205881

Protein ubiquitination regulates protein stability and modulates the composition of signaling complexes. A20 is a negative regulator of inflammatory signaling, but the molecular mechanisms involved are ill understood. Here, we generated Tnfaip3 gene-targeted A20 mutant mice bearing inactivating mutations in the zinc finger 7 (ZnF7) and ZnF4 ubiquitin-binding domains, revealing that binding to polyubiquitin is essential for A20 to suppress inflammatory disease. We demonstrate that a functional ZnF7 domain was required for recruiting A20 to the tumor necrosis factor receptor 1 (TNFR1) signaling complex and to suppress inflammatory signaling and cell death. The combined inactivation of ZnF4 and ZnF7 phenocopied the postnatal lethality and severe multiorgan inflammation of A20-deficient mice. Conditional tissue-specific expression of mutant A20 further revealed the key role of ubiquitin-binding in myeloid and intestinal epithelial cells. Collectively, these results demonstrate that the anti-inflammatory and cytoprotective functions of A20 are largely dependent on its ubiquitin-binding properties.


Inflammation/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Epithelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Polyubiquitin/metabolism , Protein Binding/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Zinc Fingers/physiology
5.
J Exp Med ; 216(9): 2010-2023, 2019 09 02.
Article En | MEDLINE | ID: mdl-31296735

The ubiquitin-editing enzyme A20 is a well-known regulator of immune cell function and homeostasis. In addition, A20 protects cells from death in an ill-defined manner. While most studies focus on its role in the TNF-receptor complex, we here identify a novel component in the A20-mediated decision between life and death. Loss of A20 in NK cells led to spontaneous NK cell death and severe NK cell lymphopenia. The few remaining NK cells showed an immature, hyperactivated phenotype, hallmarked by the basal release of cytokines and cytotoxic molecules. NK-A20-/- cells were hypersensitive to TNF-induced cell death and could be rescued, at least partially, by a combined deficiency with TNF. Unexpectedly, rapamycin, a well-established inhibitor of mTOR, also strongly protected NK-A20-/- cells from death, and further studies revealed that A20 restricts mTOR activation in NK cells. This study therefore maps A20 as a crucial regulator of mTOR signaling and underscores the need for a tightly balanced mTOR pathway in NK cell homeostasis.


Homeostasis , Killer Cells, Natural/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Survival , Lymphopenia/metabolism , Lymphopenia/pathology , Mice , Tumor Necrosis Factor alpha-Induced Protein 3/deficiency
6.
Trends Cancer ; 3(7): 491-505, 2017 07.
Article En | MEDLINE | ID: mdl-28718404

Disruption of endoplasmic reticulum (ER) homeostasis results in ER stress and activation of the unfolded protein response (UPR). This response alleviates cell stress, and is activated in both tumor cells and tumor infiltrating immune cells. The UPR plays a dual function in cancer biology, acting as a barrier to tumorigenesis at the premalignant stage, while fostering cancer maintenance in established tumors. In infiltrating immune cells, the UPR has been involved in both immunosurveillance and immunosuppressive functions. This review aims to decipher the role of the UPR at different stages of tumorigenesis and how the UPR shapes the balance between immunosurveillance and immune escape. This knowledge may improve existing UPR-targeted therapies and the design of novel strategies for cancer treatment.


Endoplasmic Reticulum Stress/immunology , Monitoring, Immunologic , Neoplasms/diagnosis , Signal Transduction/immunology , Unfolded Protein Response/immunology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinogenesis/immunology , Cell Transformation, Neoplastic/immunology , Endoplasmic Reticulum/metabolism , Humans , Immunity, Cellular/immunology , Neoplasm Staging , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Tumor Escape/immunology , Unfolded Protein Response/drug effects
7.
J Leukoc Biol ; 102(4): 1115-1126, 2017 10.
Article En | MEDLINE | ID: mdl-28716863

The prolyl hydroxylase domain-containing enzymes (PHDs) are important metabolic sensors of the cell and its environment, which might be employed to alert cells of the immune system. These enzymes regulate the expression of the hypoxia inducible factor (HIF) isoforms and NF-κB, crucial transcription factors controlling cellular metabolism and inflammation. PHD/HIF signaling is activated in the allergic lung and is proposed as a potential druggable pathway. Here, we investigated the regulation and role of the PHD isoforms in CD11c-expressing dendritic cells (DCs) and macrophages (Mϕ), sensors of the environment and crucial antigen-presenting cells in the pathogenesis of asthma. Although PHD2 and PHD3 were expressed in baseline, stimulation with house dust mite (HDM) allergen, hypoxia, and TLR4 ligands induced the expression of PHD3 in DCs. Conditional deletion or overexpression of PHD3 in CD11chi cells had minor effects on DCs and alveolar Mϕ biology in steady state. However, when put into competition with wild-type counterparts in mixed chimeric mice, alveolar Mϕ uniquely required PHD3 for optimal reconstitution of the alveolar space. Using genetic and chemical approaches, we were unable to find a clear role for PHD3 or the other PHD isoforms in DCs in asthma development. These data show cell-specific competitive advantage of PHD3 expression in antigen-presenting cells, but question whether therapeutic manipulation of PHDs in DCs would offer therapeutic benefit in asthma.


Asthma/immunology , Dendritic Cells/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Procollagen-Proline Dioxygenase/immunology , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Dendritic Cells/pathology , Gene Deletion , Lung/pathology , Macrophages, Alveolar/pathology , Mice , Mice, Transgenic , Procollagen-Proline Dioxygenase/genetics
8.
Nat Cell Biol ; 19(6): 698-710, 2017 06.
Article En | MEDLINE | ID: mdl-28459443

The IRE1-XBP1 signalling pathway is part of a cellular programme that protects against endoplasmic reticulum (ER) stress, but also controls development and survival of immune cells. Loss of XBP1 in splenic type 1 conventional dendritic cells (cDC1s) results in functional alterations without affecting cell survival. However, in mucosal cDC1s, loss of XBP1 impaired survival in a tissue-specific manner-while lung cDC1s die, intestinal cDC1s survive. This was not caused by differential activation of ER stress cell-death regulators CHOP or JNK. Rather, survival of intestinal cDC1s was associated with their ability to shut down protein synthesis through a protective integrated stress response and their marked increase in regulated IRE1-dependent messenger RNA decay. Furthermore, loss of IRE1 endonuclease on top of XBP1 led to cDC1 loss in the intestine. Thus, mucosal DCs differentially mount ATF4- and IRE1-dependent adaptive mechanisms to survive in the face of ER stress.


Dendritic Cells/enzymology , Intestinal Mucosa/enzymology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Stability , RNA, Messenger/metabolism , Respiratory Mucosa/enzymology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis , Cell Survival , Dendritic Cells/pathology , Endoplasmic Reticulum Stress , Genotype , Intestinal Mucosa/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/genetics , Mice, Transgenic , Phenotype , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Respiratory Mucosa/pathology , Signal Transduction , Time Factors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Unfolded Protein Response , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
9.
Nat Immunol ; 15(3): 248-57, 2014 Mar.
Article En | MEDLINE | ID: mdl-24441789

The role of the unfolded protein response (UPR) and endoplasmic reticulum (ER) stress in homeostasis of the immune system is incompletely understood. Here we found that dendritic cells (DCs) constitutively activated the UPR sensor IRE-1α and its target, the transcription factor XBP-1, in the absence of ER stress. Loss of XBP-1 in CD11c+ cells led to defects in phenotype, ER homeostasis and antigen presentation by CD8α+ conventional DCs, yet the closely related CD11b+ DCs were unaffected. Whereas the dysregulated ER in XBP-1-deficient DCs resulted from loss of XBP-1 transcriptional activity, the phenotypic and functional defects resulted from regulated IRE-1α-dependent degradation (RIDD) of mRNAs, including those encoding CD18 integrins and components of the major histocompatibility complex (MHC) class I machinery. Thus, a precisely regulated feedback circuit involving IRE-1α and XBP-1 controls the homeostasis of CD8α+ conventional DCs.


Cross-Priming/immunology , DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Endoribonucleases/immunology , Protein Serine-Threonine Kinases/immunology , Protein Unfolding , Transcription Factors/immunology , Unfolded Protein Response/immunology , Animals , Antigen Presentation/immunology , CD8 Antigens/immunology , CD8 Antigens/metabolism , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Endoplasmic Reticulum/immunology , Endoribonucleases/metabolism , Feedback, Physiological/physiology , Homeostasis/immunology , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/metabolism , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , X-Box Binding Protein 1
...