Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.811
Filter
Add more filters

Publication year range
1.
Cell ; 186(24): 5394-5410.e18, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37922901

ABSTRACT

Parkinson's disease (PD) is a debilitating neurodegenerative disorder. Its symptoms are typically treated with levodopa or dopamine receptor agonists, but its action lacks specificity due to the wide distribution of dopamine receptors in the central nervous system and periphery. Here, we report the development of a gene therapy strategy to selectively manipulate PD-affected circuitry. Targeting striatal D1 medium spiny neurons (MSNs), whose activity is chronically suppressed in PD, we engineered a therapeutic strategy comprised of a highly efficient retrograde adeno-associated virus (AAV), promoter elements with strong D1-MSN activity, and a chemogenetic effector to enable precise D1-MSN activation after systemic ligand administration. Application of this therapeutic approach rescues locomotion, tremor, and motor skill defects in both mouse and primate models of PD, supporting the feasibility of targeted circuit modulation tools for the treatment of PD in humans.


Subject(s)
Genetic Therapy , Parkinson Disease , Animals , Humans , Mice , Corpus Striatum/metabolism , Levodopa/therapeutic use , Levodopa/genetics , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/therapy , Primates , Receptors, Dopamine D1/metabolism , Disease Models, Animal
2.
Cell ; 184(8): 1965-1967, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33861961

ABSTRACT

Humans have an extraordinarily expanded and complex cerebral cortex, relative to non-human primates. Yet the mechanisms underlying cortical differences across evolution are unclear. A new study by Benito-Kwiecinski et al. employs cerebral organoids derived across great apes to implicate neuroepithelial progenitor shape transitions in human cortical expansion.


Subject(s)
Hominidae , Organoids , Animals , Brain , Cerebral Cortex , Primates
3.
Cell ; 184(15): 4032-4047.e31, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34171309

ABSTRACT

Although mutations in DNA are the best-studied source of neoantigens that determine response to immune checkpoint blockade, alterations in RNA splicing within cancer cells could similarly result in neoepitope production. However, the endogenous antigenicity and clinical potential of such splicing-derived epitopes have not been tested. Here, we demonstrate that pharmacologic modulation of splicing via specific drug classes generates bona fide neoantigens and elicits anti-tumor immunity, augmenting checkpoint immunotherapy. Splicing modulation inhibited tumor growth and enhanced checkpoint blockade in a manner dependent on host T cells and peptides presented on tumor MHC class I. Splicing modulation induced stereotyped splicing changes across tumor types, altering the MHC I-bound immunopeptidome to yield splicing-derived neoepitopes that trigger an anti-tumor T cell response in vivo. These data definitively identify splicing modulation as an untapped source of immunogenic peptides and provide a means to enhance response to checkpoint blockade that is readily translatable to the clinic.


Subject(s)
Neoplasms/genetics , Neoplasms/immunology , RNA Splicing/genetics , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Epitopes/immunology , Ethylenediamines/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Inflammation/pathology , Mice, Inbred C57BL , Peptides/metabolism , Protein Isoforms/metabolism , Pyrroles/pharmacology , RNA Splicing/drug effects , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
Cell ; 182(5): 1328-1340.e13, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32814014

ABSTRACT

Among arthropod vectors, ticks transmit the most diverse human and animal pathogens, leading to an increasing number of new challenges worldwide. Here we sequenced and assembled high-quality genomes of six ixodid tick species and further resequenced 678 tick specimens to understand three key aspects of ticks: genetic diversity, population structure, and pathogen distribution. We explored the genetic basis common to ticks, including heme and hemoglobin digestion, iron metabolism, and reactive oxygen species, and unveiled for the first time that genetic structure and pathogen composition in different tick species are mainly shaped by ecological and geographic factors. We further identified species-specific determinants associated with different host ranges, life cycles, and distributions. The findings of this study are an invaluable resource for research and control of ticks and tick-borne diseases.


Subject(s)
Genetic Variation/genetics , Tick-Borne Diseases/microbiology , Ticks/genetics , Animals , Cell Line , Disease Vectors , Host Specificity/genetics
5.
Cell ; 174(2): 312-324.e16, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29804838

ABSTRACT

The seven-transmembrane-spanning protein Smoothened is the central transducer in Hedgehog signaling, a pathway fundamental in development and in cancer. Smoothened is activated by cholesterol binding to its extracellular cysteine-rich domain (CRD). How this interaction leads to changes in the transmembrane domain and Smoothened activation is unknown. Here, we report crystal structures of sterol-activated Smoothened. The CRD undergoes a dramatic reorientation, allosterically causing the transmembrane domain to adopt a conformation similar to active G-protein-coupled receptors. We show that Smoothened contains a unique inhibitory π-cation lock, which is broken on activation and is disrupted in constitutively active oncogenic mutants. Smoothened activation opens a hydrophobic tunnel, suggesting a pathway for cholesterol movement from the inner membrane leaflet to the CRD. All Smoothened antagonists bind the transmembrane domain and block tunnel opening, but cyclopamine also binds the CRD, inducing the active transmembrane conformation. Together, these results define the mechanisms of Smoothened activation and inhibition.


Subject(s)
Hedgehog Proteins/metabolism , Smoothened Receptor/chemistry , Xenopus Proteins/chemistry , Allosteric Regulation , Animals , Binding Sites , Cell Line , Cholesterol/chemistry , Cholesterol/metabolism , Crystallography, X-Ray , Flow Cytometry , Hedgehog Proteins/genetics , Humans , Mice , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Protein Structure, Tertiary , Signal Transduction , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism , Veratrum Alkaloids/chemistry , Veratrum Alkaloids/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
6.
Cell ; 166(5): 1176-1187.e14, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27545348

ABSTRACT

In vertebrates, sterols are necessary for Hedgehog signaling, a pathway critical in embryogenesis and cancer. Sterols activate the membrane protein Smoothened by binding its extracellular, cysteine-rich domain (CRD). Major unanswered questions concern the nature of the endogenous, activating sterol and the mechanism by which it regulates Smoothened. We report crystal structures of CRD complexed with sterols and alone, revealing that sterols induce a dramatic conformational change of the binding site, which is sufficient for Smoothened activation and is unique among CRD-containing receptors. We demonstrate that Hedgehog signaling requires sterol binding to Smoothened and define key residues for sterol recognition and activity. We also show that cholesterol itself binds and activates Smoothened. Furthermore, the effect of oxysterols is abolished in Smoothened mutants that retain activation by cholesterol and Hedgehog. We propose that the endogenous Smoothened activator is cholesterol, not oxysterols, and that vertebrate Hedgehog signaling controls Smoothened by regulating its access to cholesterol.


Subject(s)
Cholesterol/metabolism , Hedgehog Proteins/metabolism , Smoothened Receptor/agonists , Animals , Cholesterol/chemistry , Crystallography, X-Ray , Mice , NIH 3T3 Cells , Oxysterols/chemistry , Oxysterols/metabolism , Protein Binding , Protein Conformation , Signal Transduction , Smoothened Receptor/chemistry , Smoothened Receptor/metabolism , Xenopus laevis
7.
Nat Immunol ; 19(2): 141-150, 2018 02.
Article in English | MEDLINE | ID: mdl-29292386

ABSTRACT

Cyclic diadenylate monophosphate (c-di-AMP) is secreted by bacteria as a secondary messenger. How immune cells detect c-di-AMP and initiate anti-bacterial immunity remains unknown. We found that the endoplasmic reticulum (ER) membrane adaptor ERAdP acts as a direct sensor for c-di-AMP. ERAdP-deficient mice were highly susceptible to Listeria monocytogenes infection and exhibited reduced pro-inflammatory cytokines. Mechanistically, c-di-AMP bound to the C-terminal domain of ERAdP, which in turn led to dimerization of ERAdP, resulting in association with and activation of the kinase TAK1. TAK1 activation consequently initiated activation of the transcription factor NF-κB to induce the production of pro-inflammatory cytokines in innate immune cells. Moreover, double-knockout of ERAdP and TAK1 resulted in heightened susceptibility to L. monocytogenes infection. Thus, ERAdP-mediated production of pro-inflammatory cytokines is critical for controlling bacterial infection.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Dinucleoside Phosphates/immunology , Immunity, Innate/immunology , Listeriosis/immunology , Membrane Proteins/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Second Messenger Systems/immunology
8.
Nature ; 626(7998): 347-356, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267576

ABSTRACT

To survive in a complex social group, one needs to know who to approach and, more importantly, who to avoid. In mice, a single defeat causes the losing mouse to stay away from the winner for weeks1. Here through a series of functional manipulation and recording experiments, we identify oxytocin neurons in the retrochiasmatic supraoptic nucleus (SOROXT) and oxytocin-receptor-expressing cells in the anterior subdivision of the ventromedial hypothalamus, ventrolateral part (aVMHvlOXTR) as a key circuit motif for defeat-induced social avoidance. Before defeat, aVMHvlOXTR cells minimally respond to aggressor cues. During defeat, aVMHvlOXTR cells are highly activated and, with the help of an exclusive oxytocin supply from the SOR, potentiate their responses to aggressor cues. After defeat, strong aggressor-induced aVMHvlOXTR cell activation drives the animal to avoid the aggressor and minimizes future defeat. Our study uncovers a neural process that supports rapid social learning caused by defeat and highlights the importance of the brain oxytocin system in social plasticity.


Subject(s)
Aggression , Avoidance Learning , Hypothalamus , Neural Pathways , Neurons , Oxytocin , Social Learning , Animals , Mice , Aggression/physiology , Avoidance Learning/physiology , Cues , Fear/physiology , Hypothalamus/cytology , Hypothalamus/metabolism , Neural Pathways/physiology , Neurons/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Social Behavior , Social Learning/physiology , Supraoptic Nucleus/cytology , Supraoptic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism , Neuronal Plasticity
9.
Nature ; 629(8013): 843-850, 2024 May.
Article in English | MEDLINE | ID: mdl-38658746

ABSTRACT

Angiosperms are the cornerstone of most terrestrial ecosystems and human livelihoods1,2. A robust understanding of angiosperm evolution is required to explain their rise to ecological dominance. So far, the angiosperm tree of life has been determined primarily by means of analyses of the plastid genome3,4. Many studies have drawn on this foundational work, such as classification and first insights into angiosperm diversification since their Mesozoic origins5-7. However, the limited and biased sampling of both taxa and genomes undermines confidence in the tree and its implications. Here, we build the tree of life for almost 8,000 (about 60%) angiosperm genera using a standardized set of 353 nuclear genes8. This 15-fold increase in genus-level sampling relative to comparable nuclear studies9 provides a critical test of earlier results and brings notable change to key groups, especially in rosids, while substantiating many previously predicted relationships. Scaling this tree to time using 200 fossils, we discovered that early angiosperm evolution was characterized by high gene tree conflict and explosive diversification, giving rise to more than 80% of extant angiosperm orders. Steady diversification ensued through the remaining Mesozoic Era until rates resurged in the Cenozoic Era, concurrent with decreasing global temperatures and tightly linked with gene tree conflict. Taken together, our extensive sampling combined with advanced phylogenomic methods shows the deep history and full complexity in the evolution of a megadiverse clade.


Subject(s)
Evolution, Molecular , Genes, Plant , Genomics , Magnoliopsida , Phylogeny , Fossils , Genes, Plant/genetics , Magnoliopsida/genetics , Magnoliopsida/classification , Nuclear Proteins/genetics
10.
Nature ; 617(7959): 118-124, 2023 05.
Article in English | MEDLINE | ID: mdl-37100915

ABSTRACT

Modern green revolution varieties of wheat (Triticum aestivum L.) confer semi-dwarf and lodging-resistant plant architecture owing to the Reduced height-B1b (Rht-B1b) and Rht-D1b alleles1. However, both Rht-B1b and Rht-D1b are gain-of-function mutant alleles encoding gibberellin signalling repressors that stably repress plant growth and negatively affect nitrogen-use efficiency and grain filling2-5. Therefore, the green revolution varieties of wheat harbouring Rht-B1b or Rht-D1b usually produce smaller grain and require higher nitrogen fertilizer inputs to maintain their grain yields. Here we describe a strategy to design semi-dwarf wheat varieties without the need for Rht-B1b or Rht-D1b alleles. We discovered that absence of Rht-B1 and ZnF-B (encoding a RING-type E3 ligase) through a natural deletion of a haploblock of about 500 kilobases shaped semi-dwarf plants with more compact plant architecture and substantially improved grain yield (up to 15.2%) in field trials. Further genetic analysis confirmed that the deletion of ZnF-B induced the semi-dwarf trait in the absence of the Rht-B1b and Rht-D1b alleles through attenuating brassinosteroid (BR) perception. ZnF acts as a BR signalling activator to facilitate proteasomal destruction of the BR signalling repressor BRI1 kinase inhibitor 1 (TaBKI1), and loss of ZnF stabilizes TaBKI1 to block BR signalling transduction. Our findings not only identified a pivotal BR signalling modulator but also provided a creative strategy to design high-yield semi-dwarf wheat varieties by manipulating the BR signal pathway to sustain wheat production.


Subject(s)
Biomass , Brassinosteroids , Edible Grain , Signal Transduction , Triticum , Alleles , Brassinosteroids/metabolism , Edible Grain/genetics , Edible Grain/growth & development , Edible Grain/metabolism , Gene Deletion , Genes, Plant , Gibberellins/metabolism , Phenotype , Triticum/classification , Triticum/genetics , Triticum/growth & development , Triticum/metabolism , Plant Proteins/genetics , Crops, Agricultural/genetics , Crops, Agricultural/growth & development , Crops, Agricultural/metabolism
11.
Cell ; 154(3): 623-36, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911325

ABSTRACT

The decision to initiate DNA replication is a critical step in the cell cycle of all organisms. Cells often delay replication in the face of stressful conditions, but the underlying mechanisms remain incompletely defined. Here, we demonstrate in Caulobacter crescentus that proteotoxic stress induces a cell-cycle arrest by triggering the degradation of DnaA, the conserved replication initiator. A depletion of available Hsp70 chaperone, DnaK, either through genetic manipulation or heat shock, induces synthesis of the Lon protease, which can directly degrade DnaA. Unexpectedly, we find that unfolded proteins, which accumulate following a loss of DnaK, also allosterically activate Lon to degrade DnaA, thereby ensuring a cell-cycle arrest. Our work reveals a mechanism for regulating DNA replication under adverse growth conditions. Additionally, our data indicate that unfolded proteins can actively and directly alter substrate recognition by cellular proteases.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/cytology , Caulobacter crescentus/physiology , DNA Replication , DNA-Binding Proteins/metabolism , Protease La/metabolism , Bacterial Proteins/genetics , Caulobacter crescentus/genetics , DNA, Bacterial/metabolism , DNA-Binding Proteins/genetics , Escherichia coli/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Molecular Chaperones/metabolism , Protein Folding , Sigma Factor/metabolism , Stress, Physiological
12.
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.
Nature ; 601(7892): 245-251, 2022 01.
Article in English | MEDLINE | ID: mdl-34912119

ABSTRACT

Pattern-triggered immunity (PTI) and effector-triggered immunity (ETI) in plants enable them to respond to pathogens by activating the production of defence metabolites that orchestrate immune responses1-4. How the production of defence metabolites is promoted by immune receptors and coordinated with broad-spectrum resistance remains elusive. Here we identify the deubiquitinase PICI1 as an immunity hub for PTI and ETI in rice (Oryza sativa). PICI1 deubiquitinates and stabilizes methionine synthetases to activate methionine-mediated immunity principally through biosynthesis of the phytohormone ethylene. PICI1 is targeted for degradation by blast fungal effectors, including AvrPi9, to dampen PTI. Nucleotide-binding domain, leucine-rich-repeat-containing receptors (NLRs) in the plant immune system, such as PigmR, protect PICI1 from effector-mediated degradation to reboot the methionine-ethylene cascade. Natural variation in the PICI1 gene contributes to divergence in basal blast resistance between the rice subspecies indica and japonica. Thus, NLRs govern an arms race with effectors, using a competitive mode that hinges on a critical defence metabolic pathway to synchronize PTI with ETI and ensure broad-spectrum resistance.


Subject(s)
Oryza , Plant Diseases , Methionine , Oryza/genetics , Oryza/microbiology , Plant Diseases/microbiology , Plant Immunity/genetics , Plants , Signal Transduction/genetics
15.
EMBO J ; 42(19): e112814, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37635626

ABSTRACT

The regulation of autophagy initiation is a key step in autophagosome biogenesis. However, our understanding of the molecular mechanisms underlying the stepwise assembly of ATG proteins during this process remains incomplete. The Rab GTPase Ypt1/Rab1 is recognized as an essential autophagy regulator. Here, we identify Atg23 and Atg17 as binding partners of Ypt1, with their direct interaction proving crucial for the stepwise assembly of autophagy initiation complexes. Disruption of Ypt1-Atg23 binding results in significantly reduced Atg9 interactions with Atg11, Atg13, and Atg17, thus preventing the recruitment of Atg9 vesicles to the phagophore assembly site (PAS). Likewise, Ypt1-Atg17 binding contributes to the PAS recruitment of Ypt1 and Atg1. Importantly, we found that Ypt1 is phosphorylated by TOR at the Ser174 residue. Converting this residue to alanine blocks Ypt1 phosphorylation by TOR and enhances autophagy. Conversely, the Ypt1S174D phosphorylation mimic impairs both PAS recruitment and activation of Atg1, thus inhibiting subsequent autophagy. Thus, we propose TOR-mediated Ypt1 as a multifunctional assembly factor that controls autophagy initiation via its regulation of the stepwise assembly of ATG proteins.


Subject(s)
Saccharomyces cerevisiae Proteins , Autophagy/physiology , Autophagy-Related Proteins/metabolism , Phagosomes/metabolism , Phosphorylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
16.
Genome Res ; 34(2): 189-200, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38408788

ABSTRACT

Recent studies have revealed an unexplored population of long cell-free DNA (cfDNA) molecules in human plasma using long-read sequencing technologies. However, the biological properties of long cfDNA molecules (>500 bp) remain largely unknown. To this end, we have investigated the origins of long cfDNA molecules from different genomic elements. Analysis of plasma cfDNA using long-read sequencing reveals an uneven distribution of long molecules from across the genome. Long cfDNA molecules show overrepresentation in euchromatic regions of the genome, in sharp contrast to short DNA molecules. We observe a stronger relationship between the abundance of long molecules and mRNA gene expression levels, compared with short molecules (Pearson's r = 0.71 vs. -0.14). Moreover, long and short molecules show distinct fragmentation patterns surrounding CpG sites. Leveraging the cleavage preferences surrounding CpG sites, the combined cleavage ratios of long and short molecules can differentiate patients with hepatocellular carcinoma (HCC) from non-HCC subjects (AUC = 0.87). We also investigated knockout mice in which selected nuclease genes had been inactivated in comparison with wild-type mice. The proportion of long molecules originating from transcription start sites are lower in Dffb-deficient mice but higher in Dnase1l3-deficient mice compared with that of wild-type mice. This work thus provides new insights into the biological properties and potential clinical applications of long cfDNA molecules.


Subject(s)
Carcinoma, Hepatocellular , Cell-Free Nucleic Acids , Liver Neoplasms , Humans , Animals , Mice , Cell-Free Nucleic Acids/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , DNA/genetics , Genomics , Mice, Knockout , Endodeoxyribonucleases/genetics
17.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38940293

ABSTRACT

Generation of hematopoietic stem and progenitor cells (HSPCs) ex vivo and in vivo, especially the generation of safe therapeutic HSPCs, still remains inefficient. In this study, we have identified compound BF170 hydrochloride as a previously unreported pro-hematopoiesis molecule, using the differentiation assays of primary zebrafish blastomere cell culture and mouse embryoid bodies (EBs), and we demonstrate that BF170 hydrochloride promoted definitive hematopoiesis in vivo. During zebrafish definitive hematopoiesis, BF170 hydrochloride increases blood flow, expands hemogenic endothelium (HE) cells and promotes HSPC emergence. Mechanistically, the primary cilia-Ca2+-Notch/NO signaling pathway, which is downstream of the blood flow, mediated the effects of BF170 hydrochloride on HSPC induction in vivo. Our findings, for the first time, reveal that BF170 hydrochloride is a compound that enhances HSPC induction and may be applied to the ex vivo expansion of HSPCs.


Subject(s)
Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells , Zebrafish , Animals , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Cell Differentiation/drug effects , Hematopoiesis/drug effects , Receptors, Notch/metabolism , Signal Transduction/drug effects , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Cilia/metabolism , Cilia/drug effects , Blastomeres/cytology , Blastomeres/metabolism , Blastomeres/drug effects , Cells, Cultured
18.
Nat Immunol ; 16(5): 458-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25799126

ABSTRACT

Mitophagy is essential for cellular homeostasis, but how mitophagy is regulated is largely unknown. Here we found that the kinase Jnk2 was required for stress-induced mitophagy. Jnk2 promoted ubiquitination and proteasomal degradation of the small mitochondrial form of the tumor suppressor ARF (smARF). Loss of Jnk2 led to the accumulation of smARF, which induced excessive autophagy that resulted in lysosomal degradation of the mitophagy adaptor p62 at steady state. Depletion of p62 prevented Jnk2-deficient cells from mounting mitophagy upon stress. Jnk2-deficient mice displayed defective mitophagy, which resulted in tissue damage under hypoxic stress, as well as hyperactivation of inflammasomes and increased mortality in sepsis. Our findings define a unique mechanism of maintaining immunological homeostasis that protects the host from tissue damage and mortality.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Hypoxia/immunology , Mitogen-Activated Protein Kinase 9/metabolism , Proteasome Endopeptidase Complex/metabolism , Sepsis/immunology , Animals , Cells, Cultured , DNA Damage/physiology , Female , Inflammasomes/metabolism , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Mitophagy/genetics , Proteolysis , Proto-Oncogene Proteins c-myc/metabolism , Sepsis/chemically induced , Ubiquitination
19.
Immunity ; 49(4): 740-753.e7, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30314759

ABSTRACT

Caspase-11, a cytosolic endotoxin (lipopolysaccharide: LPS) receptor, mediates pyroptosis, a lytic form of cell death. Caspase-11-dependent pyroptosis mediates lethality in endotoxemia, but it is unclear how LPS is delivered into the cytosol for the activation of caspase-11. Here we discovered that hepatocyte-released high mobility group box 1 (HMGB1) was required for caspase-11-dependent pyroptosis and lethality in endotoxemia and bacterial sepsis. Mechanistically, hepatocyte-released HMGB1 bound LPS and targeted its internalization into the lysosomes of macrophages and endothelial cells via the receptor for advanced glycation end-products (RAGE). Subsequently, HMGB1 permeabilized the phospholipid bilayer in the acidic environment of lysosomes. This resulted in LPS leakage into the cytosol and caspase-11 activation. Depletion of hepatocyte HMGB1, inhibition of hepatocyte HMGB1 release, neutralizing extracellular HMGB1, or RAGE deficiency prevented caspase-11-dependent pyroptosis and death in endotoxemia and bacterial sepsis. These findings indicate that HMGB1 interacts with LPS to mediate caspase-11-dependent pyroptosis in lethal sepsis.


Subject(s)
Caspases/immunology , Endotoxins/immunology , HMGB1 Protein/immunology , Pyroptosis/immunology , Sepsis/immunology , Animals , Caspases/genetics , Caspases/metabolism , Cells, Cultured , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endotoxins/metabolism , HEK293 Cells , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor for Advanced Glycation End Products/immunology , Receptor for Advanced Glycation End Products/metabolism , Sepsis/genetics , Sepsis/metabolism , THP-1 Cells
20.
Cell ; 150(5): 895-908, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22939619

ABSTRACT

Mutant huntingtin (HTT) protein causes Huntington disease (HD), an incurable neurological disorder. Silencing mutant HTT using nucleic acids would eliminate the root cause of HD. Developing nucleic acid drugs is challenging, and an ideal clinical approach to gene silencing would combine the simplicity of single-stranded antisense oligonucleotides with the efficiency of RNAi. Here, we describe RNAi by single-stranded siRNAs (ss-siRNAs). ss-siRNAs are potent (>100-fold more than unmodified RNA) and allele-selective (>30-fold) inhibitors of mutant HTT expression in cells derived from HD patients. Strategic placement of mismatched bases mimics micro-RNA recognition and optimizes discrimination between mutant and wild-type alleles. ss-siRNAs require Argonaute protein and function through the RNAi pathway. Intraventricular infusion of ss-siRNA produced selective silencing of the mutant HTT allele throughout the brain in a mouse HD model. These data demonstrate that chemically modified ss-siRNAs function through the RNAi pathway and provide allele-selective compounds for clinical development.


Subject(s)
Disease Models, Animal , Huntington Disease/genetics , Huntington Disease/therapy , Nerve Tissue Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Animals , Brain/metabolism , Cell Line , Humans , Huntingtin Protein , Mice , Oligodeoxyribonucleotides, Antisense/genetics , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL