Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21.869
Filter
Add more filters

Publication year range
1.
Cell ; 186(22): 4773-4787.e12, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37806310

ABSTRACT

Pollen-pistil interactions establish interspecific/intergeneric pre-zygotic hybridization barriers in plants. The rejection of undesired pollen at the stigma is crucial to avoid outcrossing but can be overcome with the support of mentor pollen. The mechanisms underlying this hybridization barrier are largely unknown. Here, in Arabidopsis, we demonstrate that receptor-like kinases FERONIA/CURVY1/ANJEA/HERCULES RECEPTOR KINASE 1 and cell wall proteins LRX3/4/5 interact on papilla cell surfaces with autocrine stigmatic RALF1/22/23/33 peptide ligands (sRALFs) to establish a lock that blocks the penetration of undesired pollen tubes. Compatible pollen-derived RALF10/11/12/13/25/26/30 peptides (pRALFs) act as a key, outcompeting sRALFs and enabling pollen tube penetration. By treating Arabidopsis stigmas with synthetic pRALFs, we unlock the barrier, facilitating pollen tube penetration from distantly related Brassicaceae species and resulting in interspecific/intergeneric hybrid embryo formation. Therefore, we uncover a "lock-and-key" system governing the hybridization breadth of interspecific/intergeneric crosses in Brassicaceae. Manipulating this system holds promise for facilitating broad hybridization in crops.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Peptide Hormones , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Brassicaceae/genetics , Brassicaceae/metabolism , Peptide Hormones/metabolism , Peptides/metabolism , Pollen/metabolism , Pollen Tube/metabolism , Reproductive Isolation
2.
Cell ; 185(17): 3138-3152.e20, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35926506

ABSTRACT

Oakleaf butterflies in the genus Kallima have a polymorphic wing phenotype, enabling these insects to masquerade as dead leaves. This iconic example of protective resemblance provides an interesting evolutionary paradigm that can be employed to study biodiversity. We integrated multi-omic data analyses and functional validation to infer the evolutionary history of Kallima species and investigate the genetic basis of their variable leaf wing patterns. We find that Kallima butterflies diversified in the eastern Himalayas and dispersed to East and Southeast Asia. Moreover, we find that leaf wing polymorphism is controlled by the wing patterning gene cortex, which has been maintained in Kallima by long-term balancing selection. Our results provide macroevolutionary and microevolutionary insights into a model species originating from a mountain ecosystem.


Subject(s)
Butterflies , Animals , Biodiversity , Biological Evolution , Butterflies/genetics , Ecosystem , Phenotype , Wings, Animal
3.
Nat Immunol ; 25(1): 102-116, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38012418

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapies have successfully treated hematological malignancies. Macrophages have also gained attention as an immunotherapy owing to their immunomodulatory capacity and ability to infiltrate solid tumors and phagocytize tumor cells. The first-generation CD3ζ-based CAR-macrophages could phagocytose tumor cells in an antigen-dependent manner. Here we engineered induced pluripotent stem cell-derived macrophages (iMACs) with toll-like receptor 4 intracellular toll/IL-1R (TIR) domain-containing CARs resulting in a markedly enhanced antitumor effect over first-generation CAR-macrophages. Moreover, the design of a tandem CD3ζ-TIR dual signaling CAR endows iMACs with both target engulfment capacity and antigen-dependent M1 polarization and M2 resistance in a nuclear factor kappa B (NF-κB)-dependent manner, as well as the capacity to modulate the tumor microenvironment. We also outline a mechanism of tumor cell elimination by CAR-induced efferocytosis against tumor cell apoptotic bodies. Taken together, we provide a second-generation CAR-iMAC with an ability for orthogonal phagocytosis and polarization and superior antitumor functions in treating solid tumors relative to first-generation CAR-macrophages.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Antigen, T-Cell , T-Lymphocytes , Cell Line, Tumor , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , Macrophages/pathology , Tumor Microenvironment
4.
Cell ; 184(12): 3178-3191.e18, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34022140

ABSTRACT

Gasdermin B (GSDMB) belongs to a large family of pore-forming cytolysins that execute inflammatory cell death programs. While genetic studies have linked GSDMB polymorphisms to human disease, its function in the immunological response to pathogens remains poorly understood. Here, we report a dynamic host-pathogen conflict between GSDMB and the IpaH7.8 effector protein secreted by enteroinvasive Shigella flexneri. We show that IpaH7.8 ubiquitinates and targets GSDMB for 26S proteasome destruction. This virulence strategy protects Shigella from the bacteriocidic activity of natural killer cells by suppressing granzyme-A-mediated activation of GSDMB. In contrast to the canonical function of most gasdermin family members, GSDMB does not inhibit Shigella by lysing host cells. Rather, it exhibits direct microbiocidal activity through recognition of phospholipids found on Gram-negative bacterial membranes. These findings place GSDMB as a central executioner of intracellular bacterial killing and reveal a mechanism employed by pathogens to counteract this host defense system.


Subject(s)
Biomarkers, Tumor/metabolism , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Neoplasm Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Shigella flexneri/physiology , Ubiquitination , Animals , Bacterial Proteins/metabolism , Cardiolipins/metabolism , Cell Line , Cell Membrane/metabolism , Female , Granzymes/metabolism , Humans , Lipid A/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microbial Viability , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Substrate Specificity
5.
Cell ; 184(9): 2362-2371.e9, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33735608

ABSTRACT

The 501Y.V2 variants of SARS-CoV-2 containing multiple mutations in spike are now dominant in South Africa and are rapidly spreading to other countries. Here, experiments with 18 pseudotyped viruses showed that the 501Y.V2 variants do not confer increased infectivity in multiple cell types except for murine ACE2-overexpressing cells, where a substantial increase in infectivity was observed. Notably, the susceptibility of the 501Y.V2 variants to 12 of 17 neutralizing monoclonal antibodies was substantially diminished, and the neutralization ability of the sera from convalescent patients and immunized mice was also reduced for these variants. The neutralization resistance was mainly caused by E484K and N501Y mutations in the receptor-binding domain of spike. The enhanced infectivity in murine ACE2-overexpressing cells suggests the possibility of spillover of the 501Y.V2 variants to mice. Moreover, the neutralization resistance we detected for the 501Y.V2 variants suggests the potential for compromised efficacy of monoclonal antibodies and vaccines.


Subject(s)
COVID-19/immunology , COVID-19/virology , Immune Evasion , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Cell Line, Tumor , HEK293 Cells , Humans , Mutation/genetics , SARS-CoV-2/genetics
6.
Nat Immunol ; 24(4): 612-624, 2023 04.
Article in English | MEDLINE | ID: mdl-36928415

ABSTRACT

Gamma delta (γδ) T cells reside within human tissues including tumors, but their function in mediating antitumor responses to immune checkpoint inhibition is unknown. Here we show that kidney cancers are infiltrated by Vδ2- γδ T cells, with equivalent representation of Vδ1+ and Vδ1- cells, that are distinct from γδ T cells found in normal human tissues. These tumor-resident Vδ2- T cells can express the transcriptional program of exhausted αß CD8+ T cells as well as canonical markers of terminal T-cell exhaustion including PD-1, TIGIT and TIM-3. Although Vδ2- γδ T cells have reduced IL-2 production, they retain expression of cytolytic effector molecules and co-stimulatory receptors such as 4-1BB. Exhausted Vδ2- γδ T cells are composed of three distinct populations that lack TCF7, are clonally expanded and express cytotoxic molecules and multiple Vδ2- T-cell receptors. Human tumor-derived Vδ2- γδ T cells maintain cytotoxic function and pro-inflammatory cytokine secretion in vitro. The transcriptional program of Vδ2- T cells in pretreatment tumor biopsies was used to predict subsequent clinical responses to PD-1 blockade in patients with cancer. Thus, Vδ2- γδ T cells within the tumor microenvironment can contribute to antitumor efficacy.


Subject(s)
CD8-Positive T-Lymphocytes , Kidney Neoplasms , Humans , CD8-Positive T-Lymphocytes/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Programmed Cell Death 1 Receptor/metabolism , Kidney Neoplasms/metabolism , T-Lymphocyte Subsets , Tumor Microenvironment
7.
Cell ; 182(5): 1284-1294.e9, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32730807

ABSTRACT

The spike protein of SARS-CoV-2 has been undergoing mutations and is highly glycosylated. It is critically important to investigate the biological significance of these mutations. Here, we investigated 80 variants and 26 glycosylation site modifications for the infectivity and reactivity to a panel of neutralizing antibodies and sera from convalescent patients. D614G, along with several variants containing both D614G and another amino acid change, were significantly more infectious. Most variants with amino acid change at receptor binding domain were less infectious, but variants including A475V, L452R, V483A, and F490L became resistant to some neutralizing antibodies. Moreover, the majority of glycosylation deletions were less infectious, whereas deletion of both N331 and N343 glycosylation drastically reduced infectivity, revealing the importance of glycosylation for viral infectivity. Interestingly, N234Q was markedly resistant to neutralizing antibodies, whereas N165Q became more sensitive. These findings could be of value in the development of vaccine and therapeutic antibodies.


Subject(s)
Antigens, Viral/genetics , Betacoronavirus/pathogenicity , Mutation , Spike Glycoprotein, Coronavirus/genetics , A549 Cells , Animals , Antigens, Viral/immunology , Betacoronavirus/genetics , Betacoronavirus/immunology , Binding Sites , Cattle , Chlorocebus aethiops , Cricetinae , Dogs , Glycosylation , HEK293 Cells , HeLa Cells , Humans , Macaca mulatta , Madin Darby Canine Kidney Cells , Mice , RAW 264.7 Cells , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Swine , Vero Cells , Virulence/genetics
8.
Cell ; 183(7): 1867-1883.e26, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33248023

ABSTRACT

Biliary atresia (BA) is a severe cholangiopathy that leads to liver failure in infants, but its pathogenesis remains to be fully characterized. By single-cell RNA profiling, we observed macrophage hypo-inflammation, Kupffer cell scavenger function defects, cytotoxic T cell expansion, and deficiency of CX3CR1+effector T and natural killer (NK) cells in infants with BA. More importantly, we discovered that hepatic B cell lymphopoiesis did not cease after birth and that tolerance defects contributed to immunoglobulin G (IgG)-autoantibody accumulation in BA. In a rhesus-rotavirus induced BA model, depleting B cells or blocking antigen presentation ameliorated liver damage. In a pilot clinical study, we demonstrated that rituximab was effective in depleting hepatic B cells and restoring the functions of macrophages, Kupffer cells, and T cells to levels comparable to those of control subjects. In summary, our comprehensive immune profiling in infants with BA had educed that B-cell-modifying therapies may alleviate liver pathology.


Subject(s)
Biliary Atresia/immunology , Biliary Atresia/therapy , Liver/immunology , Animals , Antigens, CD20/metabolism , B-Lymphocytes/immunology , Biliary Atresia/blood , Biliary Atresia/drug therapy , Biopsy , CX3C Chemokine Receptor 1/metabolism , Cell Death , Cell Line , Cell Proliferation , Cell Transdifferentiation , Child , Child, Preschool , Cohort Studies , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Humans , Immunoglobulin G/metabolism , Infant , Inflammation/pathology , Killer Cells, Natural/immunology , Kupffer Cells/pathology , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Lymphocyte Depletion , Lymphopoiesis , Male , Mice, Inbred BALB C , Phagocytosis , RNA/metabolism , Rituximab/administration & dosage , Rituximab/pharmacology , Rituximab/therapeutic use , Rotavirus/physiology , Single-Cell Analysis , Th1 Cells/immunology , Th17 Cells/immunology
9.
Cell ; 179(5): 1160-1176.e24, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730855

ABSTRACT

Pediatric-onset colitis and inflammatory bowel disease (IBD) have significant effects on the growth of infants and children, but the etiopathogenesis underlying disease subtypes remains incompletely understood. Here, we report single-cell clustering, immune phenotyping, and risk gene analysis for children with undifferentiated colitis, Crohn's disease, and ulcerative colitis. We demonstrate disease-specific characteristics, as well as common pathogenesis marked by impaired cyclic AMP (cAMP)-response signaling. Specifically, infiltration of PDE4B- and TNF-expressing macrophages, decreased abundance of CD39-expressing intraepithelial T cells, and platelet aggregation and release of 5-hydroxytryptamine at the colonic mucosae were common in colitis and IBD patients. Targeting these pathways by using the phosphodiesterase inhibitor dipyridamole restored immune homeostasis and improved colitis symptoms in a pilot study. In summary, comprehensive analysis of the colonic mucosae has uncovered common pathogenesis and therapeutic targets for children with colitis and IBD.


Subject(s)
Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/pathology , Antigens, CD/metabolism , Apyrase/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Death/drug effects , Cellular Microenvironment/drug effects , Child , Cohort Studies , Colon/pathology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dipyridamole/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Genetic Predisposition to Disease , Homeostasis/drug effects , Humans , Immunoglobulin G/blood , Immunologic Memory , Inflammation/pathology , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/genetics , Interferon Type I/metabolism , Macrophages/drug effects , Macrophages/metabolism , Methylprednisolone/pharmacology , Myeloid Cells/drug effects , Myeloid Cells/metabolism
10.
Cell ; 174(3): 758-769.e9, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30033370

ABSTRACT

While mutations affecting protein-coding regions have been examined across many cancers, structural variants at the genome-wide level are still poorly defined. Through integrative deep whole-genome and -transcriptome analysis of 101 castration-resistant prostate cancer metastases (109X tumor/38X normal coverage), we identified structural variants altering critical regulators of tumorigenesis and progression not detectable by exome approaches. Notably, we observed amplification of an intergenic enhancer region 624 kb upstream of the androgen receptor (AR) in 81% of patients, correlating with increased AR expression. Tandem duplication hotspots also occur near MYC, in lncRNAs associated with post-translational MYC regulation. Classes of structural variations were linked to distinct DNA repair deficiencies, suggesting their etiology, including associations of CDK12 mutation with tandem duplications, TP53 inactivation with inverted rearrangements and chromothripsis, and BRCA2 inactivation with deletions. Together, these observations provide a comprehensive view of how structural variations affect critical regulators in metastatic prostate cancer.


Subject(s)
Genomic Structural Variation/genetics , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , BRCA2 Protein/metabolism , Cyclin-Dependent Kinases/metabolism , DNA Copy Number Variations , Exome , Gene Expression Profiling/methods , Genomics/methods , Humans , Male , Middle Aged , Mutation , Neoplasm Metastasis/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Tandem Repeat Sequences/genetics , Tumor Suppressor Protein p53/metabolism , Whole Genome Sequencing/methods
11.
Mol Cell ; 84(3): 596-610.e6, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38215754

ABSTRACT

Although DNA N6-methyl-deoxyadenosine (6mA) is abundant in bacteria and protists, its presence and function in mammalian genomes have been less clear. We present Direct-Read 6mA sequencing (DR-6mA-seq), an antibody-independent method, to measure 6mA at base resolution. DR-6mA-seq employs a unique mutation-based strategy to reveal 6mA sites as misincorporation signatures without any chemical or enzymatic modulation of 6mA. We validated DR-6mA-seq through the successful mapping of the well-characterized G(6mA)TC motif in the E. coli DNA. As expected, when applying DR-6mA-seq to mammalian systems, we found that genomic DNA (gDNA) 6mA abundance is generally low in most mammalian tissues and cells; however, we did observe distinct gDNA 6mA sites in mouse testis and glioblastoma cells. DR-6mA-seq provides an enabling tool to detect 6mA at single-base resolution for a comprehensive understanding of DNA 6mA in eukaryotes.


Subject(s)
DNA Methylation , Escherichia coli , Animals , Mice , Escherichia coli/genetics , Genome/genetics , DNA/metabolism , Eukaryota/genetics , Deoxyadenosines/genetics , Mammals/metabolism
12.
Mol Cell ; 84(12): 2320-2336.e6, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906115

ABSTRACT

2'-O-methylation (Nm) is a prominent RNA modification well known in noncoding RNAs and more recently also found at many mRNA internal sites. However, their function and base-resolution stoichiometry remain underexplored. Here, we investigate the transcriptome-wide effect of internal site Nm on mRNA stability. Combining nanopore sequencing with our developed machine learning method, NanoNm, we identify thousands of Nm sites on mRNAs with a single-base resolution. We observe a positive effect of FBL-mediated Nm modification on mRNA stability and expression level. Elevated FBL expression in cancer cells is associated with increased expression levels for 2'-O-methylated mRNAs of cancer pathways, implying the role of FBL in post-transcriptional regulation. Lastly, we find that FBL-mediated 2'-O-methylation connects to widespread 3' UTR shortening, a mechanism that globally increases RNA stability. Collectively, we demonstrate that FBL-mediated Nm modifications at mRNA internal sites regulate gene expression by enhancing mRNA stability.


Subject(s)
3' Untranslated Regions , RNA Stability , RNA, Messenger , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Methylation , RNA Processing, Post-Transcriptional , Nanopore Sequencing/methods , Transcriptome , Gene Expression Regulation, Neoplastic , Machine Learning
13.
Nat Immunol ; 20(7): 879-889, 2019 07.
Article in English | MEDLINE | ID: mdl-31182807

ABSTRACT

CD8+ T cells and natural killer (NK) cells are central cellular components of immune responses against pathogens and cancer, which rely on interleukin (IL)-15 for homeostasis. Here we show that IL-15 also mediates homeostatic priming of CD8+ T cells for antigen-stimulated activation, which is controlled by a deubiquitinase, Otub1. IL-15 mediates membrane recruitment of Otub1, which inhibits ubiquitin-dependent activation of AKT, a kinase that is pivotal for T cell activation and metabolism. Otub1 deficiency in mice causes aberrant responses of CD8+ T cells to IL-15, rendering naive CD8+ T cells hypersensitive to antigen stimulation characterized by enhanced metabolic reprograming and effector functions. Otub1 also controls the maturation and activation of NK cells. Deletion of Otub1 profoundly enhances anticancer immunity by unleashing the activity of CD8+ T cells and NK cells. These findings suggest that Otub1 controls the activation of CD8+ T cells and NK cells by functioning as a checkpoint of IL-15-mediated priming.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cysteine Endopeptidases/metabolism , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cysteine Endopeptidases/deficiency , Deubiquitinating Enzymes/metabolism , Disease Models, Animal , Energy Metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interleukin-15/genetics , Melanoma, Experimental , Mice , Mice, Transgenic , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-15/metabolism , Self Tolerance/genetics , Self Tolerance/immunology , Signal Transduction , T-Cell Antigen Receptor Specificity , Ubiquitination
15.
Nature ; 629(8013): 893-900, 2024 May.
Article in English | MEDLINE | ID: mdl-38632402

ABSTRACT

The blood-brain barrier (BBB) protects the central nervous system from infections or harmful substances1; its impairment can lead to or exacerbate various diseases of the central nervous system2-4. However, the mechanisms of BBB disruption during infection and inflammatory conditions5,6 remain poorly defined. Here we find that activation of the pore-forming protein GSDMD by the cytosolic lipopolysaccharide (LPS) sensor caspase-11 (refs. 7-9), but not by TLR4-induced cytokines, mediates BBB breakdown in response to circulating LPS or during LPS-induced sepsis. Mice deficient in the LBP-CD14 LPS transfer and internalization pathway10-12 resist BBB disruption. Single-cell RNA-sequencing analysis reveals that brain endothelial cells (bECs), which express high levels of GSDMD, have a prominent response to circulating LPS. LPS acting on bECs primes Casp11 and Cd14 expression and induces GSDMD-mediated plasma membrane permeabilization and pyroptosis in vitro and in mice. Electron microscopy shows that this features ultrastructural changes in the disrupted BBB, including pyroptotic endothelia, abnormal appearance of tight junctions and vasculature detachment from the basement membrane. Comprehensive mouse genetic analyses, combined with a bEC-targeting adeno-associated virus system, establish that GSDMD activation in bECs underlies BBB disruption by LPS. Delivery of active GSDMD into bECs bypasses LPS stimulation and opens the BBB. In CASP4-humanized mice, Gram-negative Klebsiella pneumoniae infection disrupts the BBB; this is blocked by expression of a GSDMD-neutralizing nanobody in bECs. Our findings outline a mechanism for inflammatory BBB breakdown, and suggest potential therapies for diseases of the central nervous system associated with BBB impairment.


Subject(s)
Blood-Brain Barrier , Brain , Endothelial Cells , Gasdermins , Inflammation , Animals , Female , Humans , Male , Mice , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Blood-Brain Barrier/virology , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Caspases, Initiator/metabolism , Dependovirus , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gasdermins/antagonists & inhibitors , Gasdermins/metabolism , Inflammation/pathology , Inflammation/metabolism , Klebsiella pneumoniae/physiology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/blood , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Pyroptosis , Sepsis/metabolism , Sepsis/pathology , Sepsis/microbiology , Single-Cell Analysis , Tight Junctions/metabolism , Tight Junctions/ultrastructure
16.
Nature ; 632(8026): 782-787, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39143208

ABSTRACT

Hot-carrier transistors are a class of devices that leverage the excess kinetic energy of carriers. Unlike regular transistors, which rely on steady-state carrier transport, hot-carrier transistors modulate carriers to high-energy states, resulting in enhanced device speed and functionality. These characteristics are essential for applications that demand rapid switching and high-frequency operations, such as advanced telecommunications and cutting-edge computing technologies1-5. However, the traditional mechanisms of hot-carrier generation are either carrier injection6-11 or acceleration12,13, which limit device performance in terms of power consumption and negative differential resistance14-17. Mixed-dimensional devices, which combine bulk and low-dimensional materials, can offer different mechanisms for hot-carrier generation by leveraging the diverse potential barriers formed by energy-band combinations18-21. Here we report a hot-emitter transistor based on double mixed-dimensional graphene/germanium Schottky junctions that uses stimulated emission of heated carriers to achieve a subthreshold swing lower than 1 millivolt per decade beyond the Boltzmann limit and a negative differential resistance with a peak-to-valley current ratio greater than 100 at room temperature. Multi-valued logic with a high inverter gain and reconfigurable logic states are further demonstrated. This work reports a multifunctional hot-emitter transistor with significant potential for low-power and negative-differential-resistance applications, marking a promising advancement for the post-Moore era.

17.
Nature ; 632(8024): 301-306, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048825

ABSTRACT

Molecule-based selective contacts have become a crucial component to ensure high-efficiency inverted perovskite solar cells1-5. These molecules always consist of a conjugated core with heteroatom substitution to render the desirable carrier-transport capability6-9. So far, the design of successful conjugation cores has been limited to two N-substituted π-conjugated structures, carbazole and triphenylamine, with molecular optimization evolving around their derivatives2,5,10-12. However, further improvement of the device longevity has been hampered by the concomitant limitations of the molecular stability induced by such heteroatom-substituted structures13,14. A more robust molecular contact without sacrificing the electronic properties is in urgent demand, but remains a challenge. Here we report a peri-fused polyaromatic core structure without heteroatom substitution that yields superior carrier transport and selectivity over conventional heteroatom-substituted core structures. This core structure produced a relatively chemically inert and structurally rigid molecular contact, which considerably improved the performance of perovskite solar cells in terms of both efficiency and durability. The champion device showed an efficiency up to 26.1% with greatly improved longevity under different accelerated-ageing tests.

18.
Mol Cell ; 82(3): 542-554.e6, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35081364

ABSTRACT

Non-covalent complexes of glycolytic enzymes, called metabolons, were postulated in the 1970s, but the concept has been controversial. Here we show that a c-Myc-responsive long noncoding RNA (lncRNA) that we call glycoLINC (gLINC) acts as a backbone for metabolon formation between all four glycolytic payoff phase enzymes (PGK1, PGAM1, ENO1, and PKM2) along with lactate dehydrogenase A (LDHA). The gLINC metabolon enhances glycolytic flux, increases ATP production, and enables cell survival under serine deprivation. Furthermore, gLINC overexpression in cancer cells promotes xenograft growth in mice fed a diet deprived of serine, suggesting that cancer cells employ gLINC during metabolic reprogramming. We propose that gLINC makes a functional contribution to cancer cell adaptation and provide the first example of a lncRNA-facilitated metabolon.


Subject(s)
Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Glycolysis , Membrane Proteins/metabolism , Neoplasms/enzymology , Phosphoglycerate Kinase/metabolism , Phosphoglycerate Mutase/metabolism , Phosphopyruvate Hydratase/metabolism , RNA, Long Noncoding/metabolism , Thyroid Hormones/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Membrane Proteins/genetics , Mice, Nude , Multienzyme Complexes , Neoplasms/genetics , Neoplasms/pathology , Phosphoglycerate Kinase/genetics , Phosphoglycerate Mutase/genetics , Phosphopyruvate Hydratase/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , Serine/deficiency , Thyroid Hormones/genetics , Tumor Burden , Tumor Suppressor Proteins/genetics , Thyroid Hormone-Binding Proteins
19.
Nature ; 618(7963): 151-158, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37198494

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a lethal disease notoriously resistant to therapy1,2. This is mediated in part by a complex tumour microenvironment3, low vascularity4, and metabolic aberrations5,6. Although altered metabolism drives tumour progression, the spectrum of metabolites used as nutrients by PDA remains largely unknown. Here we identified uridine as a fuel for PDA in glucose-deprived conditions by assessing how more than 175 metabolites impacted metabolic activity in 21 pancreatic cell lines under nutrient restriction. Uridine utilization strongly correlated with the expression of uridine phosphorylase 1 (UPP1), which we demonstrate liberates uridine-derived ribose to fuel central carbon metabolism and thereby support redox balance, survival and proliferation in glucose-restricted PDA cells. In PDA, UPP1 is regulated by KRAS-MAPK signalling and is augmented by nutrient restriction. Consistently, tumours expressed high UPP1 compared with non-tumoural tissues, and UPP1 expression correlated with poor survival in cohorts of patients with PDA. Uridine is available in the tumour microenvironment, and we demonstrated that uridine-derived ribose is actively catabolized in tumours. Finally, UPP1 deletion restricted the ability of PDA cells to use uridine and blunted tumour growth in immunocompetent mouse models. Our data identify uridine utilization as an important compensatory metabolic process in nutrient-deprived PDA cells, suggesting a novel metabolic axis for PDA therapy.


Subject(s)
Glucose , Pancreatic Neoplasms , Ribose , Tumor Microenvironment , Uridine , Animals , Mice , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Ribose/metabolism , Uridine/chemistry , Glucose/deficiency , Cell Division , Cell Line, Tumor , MAP Kinase Signaling System , Uridine Phosphorylase/deficiency , Uridine Phosphorylase/genetics , Uridine Phosphorylase/metabolism , Humans
20.
Mol Cell ; 81(14): 2887-2900.e5, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34171298

ABSTRACT

WhiB7 represents a distinct subclass of transcription factors in the WhiB-Like (Wbl) family, a unique group of iron-sulfur (4Fe-4S] cluster-containing proteins exclusive to the phylum of Actinobacteria. In Mycobacterium tuberculosis (Mtb), WhiB7 interacts with domain 4 of the primary sigma factor (σA4) in the RNA polymerase holoenzyme and activates genes involved in multiple drug resistance and redox homeostasis. Here, we report crystal structures of the WhiB7:σA4 complex alone and bound to its target promoter DNA at 1.55-Å and 2.6-Å resolution, respectively. These structures show how WhiB7 regulates gene expression by interacting with both σA4 and the AT-rich sequence upstream of the -35 promoter DNA via its C-terminal DNA-binding motif, the AT-hook. By combining comparative structural analysis of the two high-resolution σA4-bound Wbl structures with molecular and biochemical approaches, we identify the structural basis of the functional divergence between the two distinct subclasses of Wbl proteins in Mtb.


Subject(s)
Bacterial Proteins/metabolism , Iron-Sulfur Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Transcription Factors/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Iron-Sulfur Proteins/genetics , Mycobacterium tuberculosis/genetics , Promoter Regions, Genetic/genetics , Sigma Factor/genetics , Sigma Factor/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL