Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93.780
Filter
Add more filters

Publication year range
1.
Cell ; 187(10): 2393-2410.e14, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38653235

ABSTRACT

SARS-CoV-2 and other sarbecoviruses continue to threaten humanity, highlighting the need to characterize common mechanisms of viral immune evasion for pandemic preparedness. Cytotoxic lymphocytes are vital for antiviral immunity and express NKG2D, an activating receptor conserved among mammals that recognizes infection-induced stress ligands (e.g., MIC-A/B). We found that SARS-CoV-2 evades NKG2D recognition by surface downregulation of MIC-A/B via shedding, observed in human lung tissue and COVID-19 patient serum. Systematic testing of SARS-CoV-2 proteins revealed that ORF6, an accessory protein uniquely conserved among sarbecoviruses, was responsible for MIC-A/B downregulation via shedding. Further investigation demonstrated that natural killer (NK) cells efficiently killed SARS-CoV-2-infected cells and limited viral spread. However, inhibition of MIC-A/B shedding with a monoclonal antibody, 7C6, further enhanced NK-cell activity toward SARS-CoV-2-infected cells. Our findings unveil a strategy employed by SARS-CoV-2 to evade cytotoxic immunity, identify the culprit immunevasin shared among sarbecoviruses, and suggest a potential novel antiviral immunotherapy.


Subject(s)
COVID-19 , Immune Evasion , Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , SARS-CoV-2 , Humans , SARS-CoV-2/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , COVID-19/immunology , COVID-19/virology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Animals , Cytotoxicity, Immunologic , Down-Regulation , Lung/immunology , Lung/virology , Lung/pathology
2.
Cell ; 185(6): 1052-1064.e12, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35180380

ABSTRACT

SARS-CoV-2 infects less than 1% of cells in the human body, yet it can cause severe damage in a variety of organs. Thus, deciphering the non-cell-autonomous effects of SARS-CoV-2 infection is imperative for understanding the cellular and molecular disruption it elicits. Neurological and cognitive defects are among the least understood symptoms of COVID-19 patients, with olfactory dysfunction being their most common sensory deficit. Here, we show that both in humans and hamsters, SARS-CoV-2 infection causes widespread downregulation of olfactory receptors (ORs) and of their signaling components. This non-cell-autonomous effect is preceded by a dramatic reorganization of the neuronal nuclear architecture, which results in dissipation of genomic compartments harboring OR genes. Our data provide a potential mechanism by which SARS-CoV-2 infection alters the cellular morphology and the transcriptome of cells it cannot infect, offering insight to its systemic effects in olfaction and beyond.


Subject(s)
Anosmia , COVID-19 , Animals , Cricetinae , Down-Regulation , Humans , Receptors, Odorant , SARS-CoV-2 , Smell
3.
Cell ; 184(3): 675-688.e19, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33421369

ABSTRACT

CRISPR-Cas systems provide prokaryotes with acquired immunity against viruses and plasmids, but how these systems are regulated to prevent autoimmunity is poorly understood. Here, we show that in the S. pyogenes CRISPR-Cas system, a long-form transactivating CRISPR RNA (tracr-L) folds into a natural single guide that directs Cas9 to transcriptionally repress its own promoter (Pcas). Further, we demonstrate that Pcas serves as a critical regulatory node. De-repression causes a dramatic 3,000-fold increase in immunization rates against viruses; however, heightened immunity comes at the cost of increased autoimmune toxicity. Using bioinformatic analyses, we provide evidence that tracrRNA-mediated autoregulation is widespread in type II-A CRISPR-Cas systems. Collectively, we unveil a new paradigm for the intrinsic regulation of CRISPR-Cas systems by natural single guides, which may facilitate the frequent horizontal transfer of these systems into new hosts that have not yet evolved their own regulatory strategies.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Expression , Homeostasis/genetics , RNA, Guide, Kinetoplastida/genetics , Autoimmunity/genetics , Base Sequence , Conserved Sequence , Down-Regulation/genetics , Models, Genetic , Mutation/genetics , Operon/genetics , Promoter Regions, Genetic/genetics , Streptococcus pyogenes/genetics , Stress, Physiological/genetics , Transcription, Genetic , Transcriptional Activation/genetics
4.
Cell ; 184(21): 5465-5481.e16, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34582787

ABSTRACT

In vivo cell fate conversions have emerged as potential regeneration-based therapeutics for injury and disease. Recent studies reported that ectopic expression or knockdown of certain factors can convert resident astrocytes into functional neurons with high efficiency, region specificity, and precise connectivity. However, using stringent lineage tracing in the mouse brain, we show that the presumed astrocyte-converted neurons are actually endogenous neurons. AAV-mediated co-expression of NEUROD1 and a reporter specifically and efficiently induces reporter-labeled neurons. However, these neurons cannot be traced retrospectively to quiescent or reactive astrocytes using lineage-mapping strategies. Instead, through a retrograde labeling approach, our results reveal that endogenous neurons are the source for these viral-reporter-labeled neurons. Similarly, despite efficient knockdown of PTBP1 in vivo, genetically traced resident astrocytes were not converted into neurons. Together, our results highlight the requirement of lineage-tracing strategies, which should be broadly applied to studies of cell fate conversions in vivo.


Subject(s)
Astrocytes/cytology , Cell Differentiation , Cell Lineage , Neurons/cytology , Animals , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/pathology , Brain Injuries/pathology , Cell Line, Tumor , Cellular Reprogramming , Dependovirus/metabolism , Down-Regulation , Gene Expression Regulation , Genes, Reporter , Glial Fibrillary Acidic Protein/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Homeodomain Proteins/metabolism , Humans , Integrases/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism
5.
Cell ; 184(20): 5089-5106.e21, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34555357

ABSTRACT

Microglia are the CNS resident immune cells that react to misfolded proteins through pattern recognition receptor ligation and activation of inflammatory pathways. Here, we studied how microglia handle and cope with α-synuclein (α-syn) fibrils and their clearance. We found that microglia exposed to α-syn establish a cellular network through the formation of F-actin-dependent intercellular connections, which transfer α-syn from overloaded microglia to neighboring naive microglia where the α-syn cargo got rapidly and effectively degraded. Lowering the α-syn burden attenuated the inflammatory profile of microglia and improved their survival. This degradation strategy was compromised in cells carrying the LRRK2 G2019S mutation. We confirmed the intercellular transfer of α-syn assemblies in microglia using organotypic slice cultures, 2-photon microscopy, and neuropathology of patients. Together, these data identify a mechanism by which microglia create an "on-demand" functional network in order to improve pathogenic α-syn clearance.


Subject(s)
Cell Membrane Structures/metabolism , Microglia/metabolism , Proteolysis , alpha-Synuclein/metabolism , Actins/metabolism , Aged , Aged, 80 and over , Animals , Apoptosis , Cytoskeleton/metabolism , Down-Regulation , Female , Humans , Inflammation/genetics , Inflammation/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Male , Mice, Inbred C57BL , Microglia/pathology , Microglia/ultrastructure , Mitochondria/metabolism , Nanotubes , Protein Aggregates , Reactive Oxygen Species/metabolism , Transcriptome/genetics
6.
Cell ; 184(22): 5559-5576.e19, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34678143

ABSTRACT

Glucose consumption is generally increased in tumor cells to support tumor growth. Interestingly, we report that glycogen accumulation is a key initiating oncogenic event during liver malignant transformation. We found that glucose-6-phosphatase (G6PC) catalyzing the last step of glycogenolysis is frequently downregulated to augment glucose storage in pre-malignant cells. Accumulated glycogen undergoes liquid-liquid phase separation, which results in the assembly of the Laforin-Mst1/2 complex and consequently sequesters Hippo kinases Mst1/2 in glycogen liquid droplets to relieve their inhibition on Yap. Moreover, G6PC or another glycogenolysis enzyme-liver glycogen phosphorylase (PYGL) deficiency in both human and mice results in glycogen storage disease along with liver enlargement and tumorigenesis in a Yap-dependent manner. Consistently, elimination of glycogen accumulation abrogates liver growth and cancer incidence, whereas increasing glycogen storage accelerates tumorigenesis. Thus, we concluded that cancer-initiating cells adapt a glycogen storing mode, which blocks Hippo signaling through glycogen phase separation to augment tumor incidence.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Glycogen/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Cell Line , Disease Models, Animal , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Glucose-6-Phosphatase/metabolism , Glycogen Phosphorylase/metabolism , Hepatocyte Growth Factor/metabolism , Hippo Signaling Pathway , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Staging , Phase Transition , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proto-Oncogene Proteins/metabolism , Serine-Threonine Kinase 3/metabolism , YAP-Signaling Proteins/metabolism
7.
Cell ; 174(5): 1095-1105.e11, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30057112

ABSTRACT

Transcriptional downregulation caused by intronic triplet repeat expansions underlies diseases such as Friedreich's ataxia. This downregulation of gene expression is coupled with epigenetic changes, but the underlying mechanisms are unknown. Here, we show that an intronic GAA/TTC triplet expansion within the IIL1 gene of Arabidopsis thaliana results in accumulation of 24-nt short interfering RNAs (siRNAs) and repressive histone marks at the IIL1 locus, which in turn causes its transcriptional downregulation and an associated phenotype. Knocking down DICER LIKE-3 (DCL3), which produces 24-nt siRNAs, suppressed transcriptional downregulation of IIL1 and the triplet expansion-associated phenotype. Furthermore, knocking down additional components of the RNA-dependent DNA methylation (RdDM) pathway also suppressed both transcriptional downregulation of IIL1 and the repeat expansion-associated phenotype. Thus, our results show that triplet repeat expansions can lead to local siRNA biogenesis, which in turn downregulates transcription through an RdDM-dependent epigenetic modification.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Epigenesis, Genetic , Introns , RNA, Plant/genetics , RNA, Small Interfering/genetics , Ribonuclease III/genetics , Transcription, Genetic , DNA Methylation , DNA Polymerase beta/genetics , Down-Regulation , Gene Expression Regulation, Plant , Genes, Plant , Oligonucleotides, Antisense/genetics , Phenotype , RNA Interference , Transgenes , Trinucleotide Repeat Expansion
8.
Cell ; 173(1): 248-259.e15, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29526463

ABSTRACT

The dynamics of the chromatin regulatory landscape during human early embryogenesis remains unknown. Using DNase I hypersensitive site (DHS) sequencing, we report that the chromatin accessibility landscape is gradually established during human early embryogenesis. Interestingly, the DHSs with OCT4 binding motifs are enriched at the timing of zygotic genome activation (ZGA) in humans, but not in mice. Consistently, OCT4 contributes to ZGA in humans, but not in mice. We further find that lower CpG promoters usually establish DHSs at later stages. Similarly, younger genes tend to establish promoter DHSs and are expressed at later embryonic stages, while older genes exhibit these features at earlier stages. Moreover, our data show that human active transposons SVA and HERV-K harbor DHSs and are highly expressed in early embryos, but not in differentiated tissues. In summary, our data provide an evolutionary developmental view for understanding the regulation of gene and transposon expression.


Subject(s)
Chromatin/metabolism , Embryo, Mammalian/metabolism , Evolution, Molecular , Animals , Binding Sites , CpG Islands , DNA Methylation , DNA Transposable Elements/genetics , Deoxyribonuclease I/metabolism , Down-Regulation , Embryonic Development , Humans , Mice , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Zygote/metabolism
9.
Cell ; 165(1): 75-87, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27015308

ABSTRACT

Transcription factor (TF) binding to DNA is fundamental for gene regulation. However, it remains unknown how the dynamics of TF-DNA interactions change during cell-fate determination in vivo. Here, we use photo-activatable FCS to quantify TF-DNA binding in single cells of developing mouse embryos. In blastocysts, the TFs Oct4 and Sox2, which control pluripotency, bind DNA more stably in pluripotent than in extraembryonic cells. By contrast, in the four-cell embryo, Sox2 engages in more long-lived interactions than does Oct4. Sox2 long-lived binding varies between blastomeres and is regulated by H3R26 methylation. Live-cell tracking demonstrates that those blastomeres with more long-lived binding contribute more pluripotent progeny, and reducing H3R26 methylation decreases long-lived binding, Sox2 target expression, and pluripotent cell numbers. Therefore, Sox2-DNA binding predicts mammalian cell fate as early as the four-cell stage. More generally, we reveal the dynamic repartitioning of TFs between DNA sites driven by physiological epigenetic changes. VIDEO ABSTRACT.


Subject(s)
SOXB1 Transcription Factors/metabolism , Animals , Blastocyst/metabolism , CARD Signaling Adaptor Proteins/metabolism , DNA/metabolism , Diffusion , Down-Regulation , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/analysis , Histones/metabolism , Kinetics , Methylation , Mice , Octamer Transcription Factor-3/metabolism , Spectrometry, Fluorescence
10.
Mol Cell ; 83(5): 653-654, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36868186

ABSTRACT

Enhancers are known for their role in mediating transcriptional activation. In this issue, Vermunt et al.1 report the unexpected finding that genes can undergo a sequential transition between distinct enhancers to mediate progressive downregulation of expression.


Subject(s)
Gene Expression , Down-Regulation , Transcriptional Activation
11.
Mol Cell ; 83(23): 4352-4369.e8, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38016474

ABSTRACT

Ferroptosis is a non-apoptotic form of regulated cell death. Glutathione (GSH) peroxidase 4 (GPX4) and GSH-independent ferroptosis suppressor protein 1 (FSP1) have been identified as major defenses. Here, we uncover a protective mechanism mediated by GSH S-transferase P1 (GSTP1) by monitoring proteinomic dynamics during ferroptosis. Dramatic downregulation of GSTP1 is caused by SMURF2-mediated GSTP1 ubiquitination and degradation at early stages of ferroptosis. Intriguingly, GSTP1 acts in GPX4- and FSP1-independent manners by catalyzing GSH conjugation of 4-hydroxynonenal and detoxifying lipid hydroperoxides via selenium-independent GSH peroxidase activity. Genetic modulation of the SMURF2/GSTP1 axis or the pharmacological inhibition of GSTP1's catalytic activity sensitized tumor responses to Food and Drug Administration (FDA)-approved ferroptosis-inducing drugs both in vitro and in vivo. GSTP1 expression also confers resistance to immune checkpoint inhibitors by blunting ferroptosis. Collectively, these findings demonstrate a GPX4/FSP1-independent cellular defense mechanism against ferroptosis and suggest that targeting SMURF2/GSTP1 to sensitize cancer cells to ferroptosis has potential as an anticancer therapy.


Subject(s)
Ferroptosis , Neoplasms , United States , Ferroptosis/genetics , Ubiquitination , Down-Regulation , Glutathione , Peroxidases , Neoplasms/genetics
12.
Immunity ; 54(6): 1168-1185.e8, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34038747

ABSTRACT

Chronic inflammation plays a central role in hepatocellular carcinoma (HCC), but the contribution of hepatocytes to tumor-associated inflammation is not clear. Here, we report that the zinc finger transcription factor Miz1 restricted hepatocyte-driven inflammation to suppress HCC, independently of its transcriptional activity. Miz1 was downregulated in HCC mouse models and a substantial fraction of HCC patients. Hepatocyte-specific Miz1 deletion in mice generated a distinct sub-group of hepatocytes that produced pro-inflammatory cytokines and chemokines, which skewed the polarization of the tumor-infiltrating macrophages toward pro-inflammatory phenotypes to promote HCC. Mechanistically, Miz1 sequestrated the oncoprotein metadherin (MTDH), preventing MTDH from promoting transcription factor nuclear factor κB (NF-κB) activation. A distinct sub-group of pro-inflammatory cytokine-producing hepatocytes was also seen in a subset of HCC patients. In addition, Miz1 expression inversely correated with disease recurrence and poor prognosis in HCC patients. Our findings identify Miz1 as a tumor suppressor that prevents hepatocytes from driving inflammation in HCC.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Hepatocytes/metabolism , Inflammation/metabolism , Liver Neoplasms/metabolism , Macrophage Activation/physiology , Protein Inhibitors of Activated STAT/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Chemokines/metabolism , Down-Regulation/physiology , Female , HEK293 Cells , Hepatocytes/pathology , Humans , Inflammation/pathology , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Zinc Fingers/physiology
13.
Cell ; 162(3): 580-92, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26213385

ABSTRACT

Although it is known that the centrioles play instructive roles in pericentriolar material (PCM) assembly and that the PCM is essential for proper centriole formation, the mechanism that governs centriole-PCM interaction is poorly understood. Here, we show that ATF5 forms a characteristic 9-fold symmetrical ring structure in the inner layer of the PCM outfitting the proximal end of the mother centriole. ATF5 controls the centriole-PCM interaction in a cell-cycle- and centriole-age-dependent manner. Interaction of ATF5 with polyglutamylated tubulin (PGT) on the mother centriole and with PCNT in the PCM renders ATF5 as a required molecule in mother centriole-directed PCM accumulation and in PCM-dependent centriole formation. ATF5 depletion blocks PCM accumulation at the centrosome and causes fragmentation of centrioles, leading to the formation of multi-polar mitotic spindles and genomic instability. These data show that ATF5 is an essential structural protein that is required for the interaction between the mother centriole and the PCM.


Subject(s)
Activating Transcription Factors/metabolism , Centrioles/metabolism , Centrosome/metabolism , Cytoskeleton/metabolism , Down-Regulation , Genomic Instability , HeLa Cells , Humans , Spindle Apparatus/metabolism , Tubulin/metabolism
14.
Cell ; 162(3): 607-21, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26232227

ABSTRACT

We identified a dominant missense mutation in the SCN transcription factor Zfhx3, termed short circuit (Zfhx3(Sci)), which accelerates circadian locomotor rhythms in mice. ZFHX3 regulates transcription via direct interaction with predicted AT motifs in target genes. The mutant protein has a decreased ability to activate consensus AT motifs in vitro. Using RNA sequencing, we found minimal effects on core clock genes in Zfhx3(Sci/+) SCN, whereas the expression of neuropeptides critical for SCN intercellular signaling was significantly disturbed. Moreover, mutant ZFHX3 had a decreased ability to activate AT motifs in the promoters of these neuropeptide genes. Lentiviral transduction of SCN slices showed that the ZFHX3-mediated activation of AT motifs is circadian, with decreased amplitude and robustness of these oscillations in Zfhx3(Sci/+) SCN slices. In conclusion, by cloning Zfhx3(Sci), we have uncovered a circadian transcriptional axis that determines the period and robustness of behavioral and SCN molecular rhythms.


Subject(s)
Circadian Rhythm , Gene Expression Regulation , Homeodomain Proteins/metabolism , Neuropeptides/genetics , Suprachiasmatic Nucleus/metabolism , Amino Acid Sequence , Animals , Down-Regulation , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Nucleotide Motifs , Promoter Regions, Genetic , Sequence Alignment , Transcription, Genetic
15.
Cell ; 163(2): 394-405, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26435104

ABSTRACT

Epithelial cells are normally stably anchored, maintaining their relative positions and association with the basement membrane. Developmental rearrangements occur through cell intercalation, and cells can delaminate during epithelial-mesenchymal transitions and metastasis. We mapped the formation of lung neuroepithelial bodies (NEBs), innervated clusters of neuroendocrine/neurosensory cells within the bronchial epithelium, revealing a targeted mode of cell migration that we named "slithering," in which cells transiently lose epithelial character but remain associated with the membrane while traversing neighboring epithelial cells to reach cluster sites. Immunostaining, lineage tracing, clonal analysis, and live imaging showed that NEB progenitors, initially distributed randomly, downregulate adhesion and polarity proteins, crawling over and between neighboring cells to converge at diametrically opposed positions at bronchial branchpoints, where they reestablish epithelial structure and express neuroendocrine genes. There is little accompanying progenitor proliferation or apoptosis. Activation of the slithering program may explain why lung cancers arising from neuroendocrine cells are highly metastatic.


Subject(s)
Cell Movement , Lung/cytology , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Neuroepithelial Bodies/cytology , Animals , Cell Lineage , Down-Regulation , Epithelial-Mesenchymal Transition , Lung/embryology , Lung/metabolism , Mice , Neuroepithelial Bodies/metabolism , Stem Cells/cytology , Stem Cells/metabolism
16.
Nature ; 629(8011): 426-434, 2024 May.
Article in English | MEDLINE | ID: mdl-38658764

ABSTRACT

Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rß-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Proliferation , Dinoprostone , Interleukin-2 , Lymphocytes, Tumor-Infiltrating , Mitochondria , Signal Transduction , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dinoprostone/metabolism , Down-Regulation , Ferroptosis , Interleukin Receptor Common gamma Subunit/biosynthesis , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mitochondria/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Microenvironment/immunology
17.
Nat Immunol ; 18(6): 654-664, 2017 06.
Article in English | MEDLINE | ID: mdl-28414311

ABSTRACT

In obesity, inflammation of white adipose tissue (AT) is associated with diminished generation of beige adipocytes ('beige adipogenesis'), a thermogenic and energy-dissipating function mediated by beige adipocytes that express the uncoupling protein UCP1. Here we delineated an inflammation-driven inhibitory mechanism of beige adipogenesis in obesity that required direct adhesive interactions between macrophages and adipocytes mediated by the integrin α4 and its counter-receptor VCAM-1, respectively; expression of the latter was upregulated in obesity. This adhesive interaction reciprocally and concomitantly modulated inflammatory activation of macrophages and downregulation of UCP1 expression dependent on the kinase Erk in adipocytes. Genetic or pharmacological inactivation of the integrin α4 in mice resulted in elevated expression of UCP1 and beige adipogenesis of subcutaneous AT in obesity. Our findings, established in both mouse systems and human systems, reveal a self-sustained cycle of inflammation-driven impairment of beige adipogenesis in obesity.


Subject(s)
Adipocytes, Beige , Adipogenesis/immunology , Adipose Tissue, White/immunology , Cell Differentiation/immunology , Inflammation/immunology , Macrophages/immunology , Obesity/immunology , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Adhesion/immunology , Diet, High-Fat , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback , Female , Gene Knockdown Techniques , Humans , Immunoblotting , Integrin alpha4/genetics , Macrophages/metabolism , Male , Mice , Middle Aged , Monocytes/immunology , Obesity/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Subcutaneous Fat , T-Lymphocytes/immunology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Young Adult
18.
Immunity ; 53(2): 243-245, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814023

ABSTRACT

Chimeric antigen receptor (CAR) T cells are potent drivers of antitumor immunity, but promoting durable CAR T cell responses remains challenging. In this issue of Immunity, Li et al. (2020) show that blockade of CAR ubiquitination induces CAR recycling to the cell surface, leading to increased CAR T cell cytotoxicity and longevity by amplifying 41BB-dependent signaling and mitochondrial metabolism.


Subject(s)
Receptors, Chimeric Antigen , Cell Line, Tumor , Down-Regulation , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
19.
Immunity ; 53(2): 456-470.e6, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32758419

ABSTRACT

Clinical evidence suggests that poor persistence of chimeric antigen receptor-T cells (CAR-T) in patients limits therapeutic efficacy. Here, we designed a CAR with recyclable capability to promote in vivo persistence and to sustain antitumor activity. We showed that the engagement of tumor antigens induced rapid ubiquitination of CARs, causing CAR downmodulation followed by lysosomal degradation. Blocking CAR ubiquitination by mutating all lysines in the CAR cytoplasmic domain (CARKR) markedly repressed CAR downmodulation by inhibiting lysosomal degradation while enhancing recycling of internalized CARs back to the cell surface. Upon encountering tumor antigens, CARKR-T cells ameliorated the loss of surface CARs, which promoted their long-term killing capacity. Moreover, CARKR-T cells containing 4-1BB signaling domains displayed elevated endosomal 4-1BB signaling that enhanced oxidative phosphorylation and promoted memory T cell differentiation, leading to superior persistence in vivo. Collectively, our study provides a straightforward strategy to optimize CAR-T antitumor efficacy by redirecting CAR trafficking.


Subject(s)
Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Down-Regulation , Female , Humans , Immunologic Memory/immunology , Immunotherapy, Adoptive , Jurkat Cells , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mitochondria/immunology , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/cytology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
20.
Cell ; 157(5): 1160-74, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855951

ABSTRACT

Developmental signaling is remarkably robust to environmental variation, including temperature. For example, in ectothermic animals such as Drosophila, Notch signaling is maintained within functional limits across a wide temperature range. We combine experimental and computational approaches to show that temperature compensation of Notch signaling is achieved by an unexpected variety of endocytic-dependent routes to Notch activation which, when superimposed on ligand-induced activation, act as a robustness module. Thermal compensation arises through an altered balance of fluxes within competing trafficking routes, coupled with temperature-dependent ubiquitination of Notch. This flexible ensemble of trafficking routes supports Notch signaling at low temperature but can be switched to restrain Notch signaling at high temperature and thus compensates for the inherent temperature sensitivity of ligand-induced activation. The outcome is to extend the physiological range over which normal development can occur. Similar mechanisms may provide thermal robustness for other developmental signals.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Endocytosis , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Down-Regulation , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Signal Transduction , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL