Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 25(6): 1046-1058, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816618

ABSTRACT

The durability of an antitumor immune response is mediated in part by the persistence of progenitor exhausted CD8+ T cells (Tpex). Tpex serve as a resource for replenishing effector T cells and preserve their quantity through self-renewal. However, it is unknown how T cell receptor (TCR) engagement affects the self-renewal capacity of Tpex in settings of continued antigen exposure. Here we use a Lewis lung carcinoma model that elicits either optimal or attenuated TCR signaling in CD8+ T cells to show that formation of Tpex in tumor-draining lymph nodes and their intratumoral persistence is dependent on optimal TCR engagement. Notably, attenuated TCR stimulation accelerates the terminal differentiation of optimally primed Tpex. This TCR-reinforced Tpex development and self-renewal is coupled to proximal positioning to dendritic cells and epigenetic imprinting involving increased chromatin accessibility at Egr2 and Tcf1 target loci. Collectively, this study highlights the critical function of TCR engagement in sustaining Tpex during tumor progression.


Subject(s)
CD8-Positive T-Lymphocytes , Carcinoma, Lewis Lung , Hepatocyte Nuclear Factor 1-alpha , Mice, Inbred C57BL , Receptors, Antigen, T-Cell , Animals , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Mice , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Cell Differentiation/immunology , Dendritic Cells/immunology , Signal Transduction/immunology , Mice, Knockout , Lymphocyte Activation/immunology , Cell Self Renewal , Mice, Transgenic , Early Growth Response Protein 2
2.
Cell Rep Med ; 5(3): 101469, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508137

ABSTRACT

Fibrolamellar carcinoma (FLC) is a liver tumor with a high mortality burden and few treatment options. A promising therapeutic vulnerability in FLC is its driver mutation, a conserved DNAJB1-PRKACA gene fusion that could be an ideal target neoantigen for immunotherapy. In this study, we aim to define endogenous CD8 T cell responses to this fusion in FLC patients and evaluate fusion-specific T cell receptors (TCRs) for use in cellular immunotherapies. We observe that fusion-specific CD8 T cells are rare and that FLC patient TCR repertoires lack large clusters of related TCR sequences characteristic of potent antigen-specific responses, potentially explaining why endogenous immune responses are insufficient to clear FLC tumors. Nevertheless, we define two functional fusion-specific TCRs, one of which has strong anti-tumor activity in vivo. Together, our results provide insights into the fragmented nature of neoantigen-specific repertoires in humans and indicate routes for clinical development of successful immunotherapies for FLC.


Subject(s)
Carcinoma, Hepatocellular , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/pathology , Cell- and Tissue-Based Therapy , HSP40 Heat-Shock Proteins/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics
3.
Antioxidants (Basel) ; 12(11)2023 Nov 16.
Article in English | MEDLINE | ID: mdl-38001860

ABSTRACT

The endoplasmic reticulum is a subcellular organelle key in the control of synthesis, folding, and sorting of proteins. Under endoplasmic reticulum stress, an adaptative unfolded protein response is activated; however, if this activation is prolonged, cells can undergo cell death, in part due to oxidative stress and mitochondrial fragmentation. Here, we report that endoplasmic reticulum stress activates c-Abl tyrosine kinase, inducing its translocation to mitochondria. We found that endoplasmic reticulum stress-activated c-Abl interacts with and phosphorylates the mitochondrial fusion protein MFN2, resulting in mitochondrial fragmentation and apoptosis. Moreover, the pharmacological or genetic inhibition of c-Abl prevents MFN2 phosphorylation, mitochondrial fragmentation, and apoptosis in cells under endoplasmic reticulum stress. Finally, in the amyotrophic lateral sclerosis mouse model, where endoplasmic reticulum and oxidative stress has been linked to neuronal cell death, we demonstrated that the administration of c-Abl inhibitor neurotinib delays the onset of symptoms. Our results uncovered a function of c-Abl in the crosstalk between endoplasmic reticulum stress and mitochondrial dynamics via MFN2 phosphorylation.

4.
Nat Immunol ; 24(10): 1735-1747, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37679549

ABSTRACT

Neurodegenerative diseases, including Alzheimer's disease (AD), are characterized by innate immune-mediated inflammation, but functional and mechanistic effects of the adaptive immune system remain unclear. Here we identify brain-resident CD8+ T cells that coexpress CXCR6 and PD-1 and are in proximity to plaque-associated microglia in human and mouse AD brains. We also establish that CD8+ T cells restrict AD pathologies, including ß-amyloid deposition and cognitive decline. Ligand-receptor interaction analysis identifies CXCL16-CXCR6 intercellular communication between microglia and CD8+ T cells. Further, Cxcr6 deficiency impairs accumulation, tissue residency programming and clonal expansion of brain PD-1+CD8+ T cells. Ablation of Cxcr6 or CD8+ T cells ultimately increases proinflammatory cytokine production from microglia, with CXCR6 orchestrating brain CD8+ T cell-microglia colocalization. Collectively, our study reveals protective roles for brain CD8+ T cells and CXCR6 in mouse AD pathogenesis and highlights that microenvironment-specific, intercellular communication orchestrates tissue homeostasis and protection from neuroinflammation.

5.
Nature ; 620(7972): 200-208, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37407815

ABSTRACT

Cancer cells evade T cell-mediated killing through tumour-immune interactions whose mechanisms are not well understood1,2. Dendritic cells (DCs), especially type-1 conventional DCs (cDC1s), mediate T cell priming and therapeutic efficacy against tumours3. DC functions are orchestrated by pattern recognition receptors3-5, although other signals involved remain incompletely defined. Nutrients are emerging mediators of adaptive immunity6-8, but whether nutrients affect DC function or communication between innate and adaptive immune cells is largely unresolved. Here we establish glutamine as an intercellular metabolic checkpoint that dictates tumour-cDC1 crosstalk and licenses cDC1 function in activating cytotoxic T cells. Intratumoral glutamine supplementation inhibits tumour growth by augmenting cDC1-mediated CD8+ T cell immunity, and overcomes therapeutic resistance to checkpoint blockade and T cell-mediated immunotherapies. Mechanistically, tumour cells and cDC1s compete for glutamine uptake via the transporter SLC38A2 to tune anti-tumour immunity. Nutrient screening and integrative analyses show that glutamine is the dominant amino acid in promoting cDC1 function. Further, glutamine signalling via FLCN impinges on TFEB function. Loss of FLCN in DCs selectively impairs cDC1 function in vivo in a TFEB-dependent manner and phenocopies SLC38A2 deficiency by eliminating the anti-tumour therapeutic effect of glutamine supplementation. Our findings establish glutamine-mediated intercellular metabolic crosstalk between tumour cells and cDC1s that underpins tumour immune evasion, and reveal glutamine acquisition and signalling in cDC1s as limiting events for DC activation and putative targets for cancer treatment.


Subject(s)
Amino Acid Transport System A , Dendritic Cells , Glutamine , Neoplasms , Signal Transduction , Amino Acid Transport System A/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glutamine/metabolism , Neoplasms/immunology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism
6.
Res Sq ; 2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37333156

ABSTRACT

Understanding interactions between adoptively transferred immune cells and the tumor immune microenvironment (TIME) is critical for developing successful T-cell based immunotherapies. Here we investigated the impact of the TIME and chimeric antigen receptor (CAR) design on anti-glioma activity of B7-H3-specific CAR T-cells. We show that five out of six B7-H3 CARs with varying transmembrane, co-stimulatory, and activation domains, exhibit robust functionality in vitro. However, in an immunocompetent glioma model, these CAR T-cells demonstrated significantly varied levels of anti-tumor activity. We used single-cell RNA sequencing to examine the brain TIME after CAR T-cell therapy. We show that the TIME composition was influenced by CAR T-cell treatment. We also found that successful anti-tumor responses were supported by the presence and activity of macrophages and endogenous T-cells. Together, our study demonstrates that efficacy of CAR T-cell therapy in high-grade glioma is dependent on CAR structural design and its capacity to modulate the TIME.

7.
Sci Adv ; 9(21): eadg8156, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37224250

ABSTRACT

Degradation of defective mitochondria is an essential process to maintain cellular homeostasis and it is strictly regulated by the ubiquitin-proteasome system (UPS) and lysosomal activities. Here, using genome-wide CRISPR and small interference RNA screens, we identified a critical contribution of the lysosomal system in controlling aberrant induction of apoptosis following mitochondrial damage. After treatment with mitochondrial toxins, activation of the PINK1-Parkin axis triggered a BAX- and BAK-independent process of cytochrome c release from mitochondria followed by APAF1 and caspase 9-dependent apoptosis. This phenomenon was mediated by UPS-dependent outer mitochondrial membrane (OMM) degradation and was reversed using proteasome inhibitors. We found that the subsequent recruitment of the autophagy machinery to the OMM protected cells from apoptosis, mediating the lysosomal degradation of dysfunctional mitochondria. Our results underscore a major role of the autophagy machinery in counteracting aberrant noncanonical apoptosis and identified autophagy receptors as key elements in the regulation of this process.


Subject(s)
Apoptosis , Mitophagy , bcl-2-Associated X Protein/genetics , Autophagy , Mitochondria , Ubiquitin
8.
mBio ; 14(3): e0051023, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37052506

ABSTRACT

Microbial components have a range of direct effects on the fetal brain. However, little is known about the cellular targets and molecular mechanisms that mediate these effects. Neural progenitor cells (NPCs) control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. We identify ventricular radial glia (vRG), the primary NPC, as the target of bacterial cell wall (BCW) generated during the antibiotic treatment of maternal pneumonia. BCW enhanced proliferative potential of vRGs by shortening the cell cycle and increasing self-renewal. Expanded vRGs propagated to increase neuronal output in all cortical layers. Remarkably, Toll-like receptor 2 (TLR2), which recognizes BCW, localized at the base of primary cilia in vRGs and the BCW-TLR2 interaction suppressed ciliogenesis leading to derepression of Hedgehog (HH) signaling and expansion of vRGs. We also show that TLR6 is an essential partner of TLR2 in this process. Surprisingly, TLR6 alone was required to set the number of cortical neurons under healthy conditions. These findings suggest that an endogenous signal from TLRs suppresses cortical expansion during normal development of the neocortex and that BCW antagonizes that signal through the TLR2/cilia/HH signaling axis changing brain structure and function. IMPORTANCE Fetal brain development in early gestation can be impacted by transplacental infection, altered metabolites from the maternal microbiome, or maternal immune activation. It is less well understood how maternal microbial subcomponents that cross the placenta, such as bacterial cell wall (BCW), directly interact with fetal neural progenitors and neurons and affect development. This scenario plays out in the clinic when BCW debris released during antibiotic therapy of maternal infection traffics to the fetal brain. This study identifies the direct interaction of BCW with TLR2/6 present on the primary cilium, the signaling hub on fetal neural progenitor cells (NPCs). NPCs control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. Within a window of vulnerability before the appearance of fetal immune cells, the BCW-TLR2/6 interaction results in the inhibition of ciliogenesis, derepression of Sonic Hedgehog signaling, excess proliferation of neural progenitors, and abnormal cortical architecture. In the first example of TLR signaling linked to Sonic Hedgehog, BCW/TLR2/6 appears to act during fetal brain morphogenesis to play a role in setting the total cell number in the neocortex.


Subject(s)
Hedgehog Proteins , Neocortex , Pregnancy , Female , Humans , Hedgehog Proteins/metabolism , Neocortex/metabolism , Toll-Like Receptor 2/metabolism , Ligands , Toll-Like Receptor 6/metabolism
9.
Proc Natl Acad Sci U S A ; 119(41): e2207240119, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36191211

ABSTRACT

The absence of Caspase-8 or its adapter, Fas-associated death domain (FADD), results in activation of receptor interacting protein kinase-3 (RIPK3)- and mixed-lineage kinase-like (MLKL)-dependent necroptosis in vivo. Here, we show that spontaneous activation of RIPK3, phosphorylation of MLKL, and necroptosis in Caspase-8- or FADD-deficient cells was dependent on the nucleic acid sensor, Z-DNA binding protein-1 (ZBP1). We genetically engineered a mouse model by a single insertion of FLAG tag onto the N terminus of endogenous MLKL (MlklFLAG/FLAG), creating an inactive form of MLKL that permits monitoring of phosphorylated MLKL without activating necroptotic cell death. Casp8-/-MlklFLAG/FLAG mice were viable and displayed phosphorylated MLKL in a variety of tissues, together with dramatically increased expression of ZBP1 compared to Casp8+/+ mice. Studies in vitro revealed an increased expression of ZBP1 in cells lacking FADD or Caspase-8, which was suppressed by reconstitution of Caspase-8 or FADD. Ablation of ZBP1 in Casp8-/-MlklFLAG/FLAG mice suppressed spontaneous MLKL phosphorylation in vivo. ZBP1 expression and downstream activation of RIPK3 and MLKL in cells lacking Caspase-8 or FADD relied on a positive feedback mechanism requiring the nucleic acid sensors cyclic GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), and TBK1 signaling pathways. Our study identifies a molecular mechanism whereby Caspase-8 and FADD suppress spontaneous necroptotic cell death.


Subject(s)
Necroptosis , Nucleic Acids , Animals , Apoptosis/physiology , Caspase 8/genetics , Caspase 8/metabolism , DNA-Binding Proteins/metabolism , Fas-Associated Death Domain Protein/genetics , Interferons/metabolism , Mice , Nucleotidyltransferases/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
11.
Cell Death Differ ; 29(7): 1318-1334, 2022 07.
Article in English | MEDLINE | ID: mdl-35726022

ABSTRACT

The ability of mitochondria to buffer a rapid rise in cytosolic Ca2+ is a hallmark of proper cell homeostasis. Here, we employed m-3M3FBS, a putative phospholipase C (PLC) agonist, to explore the relationships between intracellular Ca2+ imbalance, mitochondrial physiology, and cell death. m-3M3FBS induced a potent dose-dependent Ca2+ release from the endoplasmic reticulum (ER), followed by a rise in intra-mitochondrial Ca2+. When the latter exceeded the organelle buffering capacity, an abrupt mitochondrial inner membrane permeabilization (MIMP) occurred, releasing matrix contents into the cytosol. MIMP was followed by cell death that was independent of Bcl-2 family members and inhibitable by the intracellular Ca2+ chelator BAPTA-AM. Cyclosporin A (CsA), capable of blocking the mitochondrial permeability transition (MPT), completely prevented cell death induced by m-3M3FBS. However, CsA acted upstream of mitochondria by preventing Ca2+ release from ER stores. Therefore, loss of Ca2+ intracellular balance and mitochondrial Ca2+ overload followed by MIMP induced a cell death process that is distinct from Bcl-2 family-regulated mitochondrial outer membrane permeabilization (MOMP). Further, the inhibition of cell death by CsA or its analogues can be independent of effects on the MPT.


Subject(s)
Calcium , Mitochondrial Membranes , Apoptosis , Calcium/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , bcl-2-Associated X Protein/metabolism
12.
Nature ; 600(7888): 308-313, 2021 12.
Article in English | MEDLINE | ID: mdl-34795452

ABSTRACT

Nutrients are emerging regulators of adaptive immunity1. Selective nutrients interplay with immunological signals to activate mechanistic target of rapamycin complex 1 (mTORC1), a key driver of cell metabolism2-4, but how these environmental signals are integrated for immune regulation remains unclear. Here we use genome-wide CRISPR screening combined with protein-protein interaction networks to identify regulatory modules that mediate immune receptor- and nutrient-dependent signalling to mTORC1 in mouse regulatory T (Treg) cells. SEC31A is identified to promote mTORC1 activation by interacting with the GATOR2 component SEC13 to protect it from SKP1-dependent proteasomal degradation. Accordingly, loss of SEC31A impairs T cell priming and Treg suppressive function in mice. In addition, the SWI/SNF complex restricts expression of the amino acid sensor CASTOR1, thereby enhancing mTORC1 activation. Moreover, we reveal that the CCDC101-associated SAGA complex is a potent inhibitor of mTORC1, which limits the expression of glucose and amino acid transporters and maintains T cell quiescence in vivo. Specific deletion of Ccdc101 in mouse Treg cells results in uncontrolled inflammation but improved antitumour immunity. Collectively, our results establish epigenetic and post-translational mechanisms that underpin how nutrient transporters, sensors and transducers interplay with immune signals for three-tiered regulation of mTORC1 activity and identify their pivotal roles in licensing T cell immunity and immune tolerance.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Nutrients , Protein Interaction Maps , T-Lymphocytes, Regulatory , Animals , Female , Male , Mice , Carrier Proteins/metabolism , CRISPR-Cas Systems/genetics , Forkhead Transcription Factors/metabolism , Genome/genetics , Homeostasis , Immune Tolerance , Inflammation/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/immunology , Nuclear Proteins/metabolism , Nutrients/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , S-Phase Kinase-Associated Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Trans-Activators/metabolism
13.
J Exp Med ; 217(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32289155

ABSTRACT

Invariant natural killer T (iNKT) cells acquire effector functions during development by mechanisms that remain poorly understood. Here, we show that the Hippo kinases Mst1 and Mst2 act as molecular rheostats for the terminal maturation and effector differentiation programs of iNKT cells. Loss of Mst1 alone or together with Mst2 impedes iNKT cell development, associated with defective IL-15-dependent cell survival. Mechanistically, Mst1 enforces iNKT cellular and transcriptional quiescence associated with maturation and commitment to iNKT1 cells by suppressing proliferation and Opa1-related mitochondrial metabolism that are dynamically regulated during iNKT cell development. Furthermore, Mst1 shapes the reciprocal fate decisions between iNKT1 and iNKT17 effector cells, which respectively depend upon mitochondrial dynamics and ICOS-mTORC2 signaling. Collectively, these findings establish Mst1 as a crucial regulator of mitochondrial homeostasis and quiescence in iNKT cell development and effector lineage differentiation and highlight that establishment of quiescence programs underlies iNKT cell development and effector maturation.


Subject(s)
Cell Cycle , Cell Lineage , Hepatocyte Growth Factor/metabolism , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Survival , Gene Expression Regulation , Hippo Signaling Pathway , Homeostasis , Interleukin-15/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Serine-Threonine Kinase 3 , Transcription, Genetic
14.
J Exp Med ; 217(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31722972

ABSTRACT

Thymocyte egress is a critical determinant of T cell homeostasis and adaptive immunity. Despite the roles of G protein-coupled receptors in thymocyte emigration, the downstream signaling mechanism remains poorly defined. Here, we report the discrete roles for the two branches of mevalonate metabolism-fueled protein prenylation pathway in thymocyte egress and immune homeostasis. The protein geranylgeranyltransferase Pggt1b is up-regulated in single-positive thymocytes, and loss of Pggt1b leads to marked defects in thymocyte egress and T cell lymphopenia in peripheral lymphoid organs in vivo. Mechanistically, Pggt1b bridges sphingosine-1-phosphate and chemokine-induced migratory signals with the activation of Cdc42 and Pak signaling and mevalonate-dependent thymocyte trafficking. In contrast, the farnesyltransferase Fntb, which mediates a biochemically similar process of protein farnesylation, is dispensable for thymocyte egress but contributes to peripheral T cell homeostasis. Collectively, our studies establish context-dependent effects of protein prenylation and unique roles of geranylgeranylation in thymic egress and highlight that the interplay between cellular metabolism and posttranslational modification underlies immune homeostasis.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Mevalonic Acid/metabolism , Protein Prenylation/genetics , Thymocytes/metabolism , Alkyl and Aryl Transferases/genetics , Animals , Cell Movement/genetics , Cells, Cultured , Farnesyltranstransferase/genetics , Farnesyltranstransferase/metabolism , Homeostasis/genetics , Homeostasis/immunology , Lymphopenia/genetics , Lysophospholipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , T-Lymphocytes/metabolism , Thymus Gland/immunology , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism
15.
Nature ; 576(7787): 471-476, 2019 12.
Article in English | MEDLINE | ID: mdl-31827283

ABSTRACT

Adoptive cell therapy represents a new paradigm in cancer immunotherapy, but it can be limited by the poor persistence and function of transferred T cells1. Here we use an in vivo pooled CRISPR-Cas9 mutagenesis screening approach to demonstrate that, by targeting REGNASE-1, CD8+ T cells are reprogrammed to long-lived effector cells with extensive accumulation, better persistence and robust effector function in tumours. REGNASE-1-deficient CD8+ T cells show markedly improved therapeutic efficacy against mouse models of melanoma and leukaemia. By using a secondary genome-scale CRISPR-Cas9 screening, we identify BATF as the key target of REGNASE-1 and as a rheostat that shapes antitumour responses. Loss of BATF suppresses the increased accumulation and mitochondrial fitness of REGNASE-1-deficient CD8+ T cells. By contrast, the targeting of additional signalling factors-including PTPN2 and SOCS1-improves the therapeutic efficacy of REGNASE-1-deficient CD8+ T cells. Our findings suggest that T cell persistence and effector function can be coordinated in tumour immunity and point to avenues for improving the efficacy of adoptive cell therapy for cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Leukemia/immunology , Leukemia/therapy , Melanoma/immunology , Melanoma/therapy , Molecular Targeted Therapy , Ribonucleases/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/cytology , CRISPR-Cas Systems/genetics , Disease Models, Animal , Female , Gene Deletion , Humans , Leukemia/genetics , Leukemia/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mitochondria/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Reproducibility of Results , Ribonucleases/deficiency , Ribonucleases/genetics , Ribonucleases/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Tumor Microenvironment/immunology
16.
Immunity ; 51(6): 1012-1027.e7, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31668641

ABSTRACT

Regulatory T (Treg) cells are critical mediators of immune tolerance whose activity depends upon T cell receptor (TCR) and mTORC1 kinase signaling, but the mechanisms that dictate functional activation of these pathways are incompletely understood. Here, we showed that amino acids license Treg cell function by priming and sustaining TCR-induced mTORC1 activity. mTORC1 activation was induced by amino acids, especially arginine and leucine, accompanied by the dynamic lysosomal localization of the mTOR and Tsc complexes. Rag and Rheb GTPases were central regulators of amino acid-dependent mTORC1 activation in effector Treg (eTreg) cells. Mice bearing RagA-RagB- or Rheb1-Rheb2-deficient Treg cells developed a fatal autoimmune disease and had reduced eTreg cell accumulation and function. RagA-RagB regulated mitochondrial and lysosomal fitness, while Rheb1-Rheb2 enforced eTreg cell suppressive gene signature. Together, these findings reveal a crucial requirement of amino acid signaling for licensing and sustaining mTORC1 activation and functional programming of Treg cells.


Subject(s)
Arginine/metabolism , Leucine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Cycle , Cell Differentiation/physiology , Cell Line , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Ras Homolog Enriched in Brain Protein/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/cytology
17.
J Clin Invest ; 129(6): 2251-2265, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31033479

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex autoimmune disease with genetic and environmental contributions. Hallmarks of the disease are the appearance of immune complexes (IC) containing autoreactive Abs and TLR-activating nucleic acids, whose deposition in kidney glomeruli is suspected to promote tissue injury and glomerulonephritis (GN). Here, using a mouse model based on the human SLE susceptibility locus TNFAIP3-interacting protein 1 (TNIP1, also known as ABIN1), we investigated the pathogenesis of GN. We found that GN was driven by TLRs but, remarkably, proceeded independently of ICs. Rather, disease in 3 different mouse models and patients with SLE was characterized by glomerular accumulation of patrolling monocytes (PMos), a cell type with an emerging key function in vascular inflammation. Consistent with such function in GN, monocyte-specific deletion of ABIN1 promoted kidney disease, whereas selective elimination of PMos provided protection. In contrast to GN, PMo elimination did not protect from reduced survival or disease symptoms such as IC generation and splenomegaly, suggesting that GN and other inflammatory processes are governed by distinct pathogenic mechanisms. These data identify TLR-activated PMos as the principal component of an intravascular process that contributes to glomerular inflammation and kidney injury.


Subject(s)
Lupus Nephritis/immunology , Monocytes/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Mice , Mice, Knockout , Monocytes/pathology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
18.
Cell Res ; 29(5): 391-405, 2019 05.
Article in English | MEDLINE | ID: mdl-30911060

ABSTRACT

Dendritic cells (DCs) play a pivotal role in priming adaptive immunity. However, the involvement of DCs in controlling excessive and deleterious T cell responses remains poorly defined. Moreover, the metabolic dependence and regulation of DC function are unclear. Here we show that LKB1 signaling in DCs functions as a brake to restrain excessive tumor-promoting regulatory T cell (Treg) and Th17 cell responses, thereby promoting protective anti-tumor immunity and maintaining proper immune homeostasis. LKB1 deficiency results in dysregulated metabolism and mTOR activation of DCs. Loss of LKB1 also leads to aberrant DC maturation and production of cytokines and immunoregulatory molecules. Blocking mTOR signaling in LKB1-deficient DCs partially rectifies the abnormal phenotypes of DC activation and Treg expansion, whereas uncontrolled Th17 responses depend upon IL-6-STAT3 signaling. By coordinating metabolic and immune quiescence of DCs, LKB1 acts as a crucial signaling hub in DCs to enforce protective anti-tumor immunity and normal immune homeostasis.


Subject(s)
Dendritic Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Cell Differentiation , Cell Line, Tumor , Dendritic Cells/cytology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism , Thymus Gland/metabolism
19.
Immunity ; 49(5): 899-914.e6, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413360

ABSTRACT

Interleukin-2 (IL-2) and downstream transcription factor STAT5 are important for maintaining regulatory T (Treg) cell homeostasis and function. Treg cells can respond to low IL-2 levels, but the mechanisms of STAT5 activation during partial IL-2 deficiency remain uncertain. We identified the serine-threonine kinase Mst1 as a signal-dependent amplifier of IL-2-STAT5 activity in Treg cells. High Mst1 and Mst2 (Mst1-Mst2) activity in Treg cells was crucial to prevent tumor resistance and autoimmunity. Mechanistically, Mst1-Mst2 sensed IL-2 signals to promote the STAT5 activation necessary for Treg cell homeostasis and lineage stability and to maintain the highly suppressive phosphorylated-STAT5+ Treg cell subpopulation. Unbiased quantitative proteomics revealed association of Mst1 with the cytoskeletal DOCK8-LRCHs module. Mst1 deficiency limited Treg cell migration and access to IL-2 and activity of the small GTPase Rac, which mediated downstream STAT5 activation. Collectively, IL-2-STAT5 signaling depends upon Mst1-Mst2 functions to maintain a stable Treg cell pool and immune tolerance.


Subject(s)
Hepatocyte Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Cell Lineage/genetics , Hepatocyte Growth Factor/genetics , Hippo Signaling Pathway , Interleukin-2/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Serine-Threonine Kinase 3 , T-Lymphocytes, Regulatory/immunology , rac GTP-Binding Proteins/metabolism
20.
J Vis Exp ; (138)2018 08 07.
Article in English | MEDLINE | ID: mdl-30148498

ABSTRACT

Necroptosis is a programmed cell death pathway triggered by activation of receptor interacting protein kinase 3 (RIPK3), which phosphorylates and activates the mixed lineage kinase-like domain pseudokinase, MLKL, to rupture or permeabilize the plasma membrane. Necroptosis is an inflammatory pathway associated with multiple pathologies including autoimmunity, infectious and cardiovascular diseases, stroke, neurodegeneration, and cancer. Here, we describe protocols that can be used to characterize MLKL as the executioner of plasma membrane rupture in necroptosis. We visualize the process of necroptosis in cells using live-cell imaging with conventional and confocal fluorescence microscopy, and in fixed cells using electron microscopy, which together revealed the redistribution of MLKL from the cytosol to the plasma membrane prior to induction of large holes in the plasma membrane. We present in vitro nuclear magnetic resonance (NMR) analysis using lipids to identify putative modulators of MLKL-mediated necroptosis. Based on this method, we identified quantitative lipid-binding preferences and phosphatidyl-inositol phosphates (PIPs) as critical binders of MLKL that are required for plasma membrane targeting and permeabilization in necroptosis.


Subject(s)
Necrosis/genetics , Protein Kinases/metabolism , Animals , Humans , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...