Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
1.
Nat Rev Mol Cell Biol ; 18(2): 90-101, 2017 02.
Article in English | MEDLINE | ID: mdl-27924077

ABSTRACT

Eight types of short-chain Lys acylations have recently been identified on histones: propionylation, butyrylation, 2-hydroxyisobutyrylation, succinylation, malonylation, glutarylation, crotonylation and ß-hydroxybutyrylation. Emerging evidence suggests that these histone modifications affect gene expression and are structurally and functionally different from the widely studied histone Lys acetylation. In this Review, we discuss the regulation of non-acetyl histone acylation by enzymatic and metabolic mechanisms, the acylation 'reader' proteins that mediate the effects of different acylations and their physiological functions, which include signal-dependent gene activation, spermatogenesis, tissue injury and metabolic stress. We propose a model to explain our present understanding of how differential histone acylation is regulated by the metabolism of the different acyl-CoA forms, which in turn modulates the regulation of gene expression.


Subject(s)
Gene Expression Regulation , Histones/chemistry , Histones/metabolism , Acetyl Coenzyme A/metabolism , Acyl Coenzyme A/metabolism , Acylation , Animals , Fatty Acids, Volatile/metabolism , Histones/genetics , Humans , Lysine/metabolism , Male , Protein Domains , Protein Processing, Post-Translational , Spermatogenesis , Stress, Physiological
2.
Nature ; 617(7962): 818-826, 2023 05.
Article in English | MEDLINE | ID: mdl-37198486

ABSTRACT

Cancer cells rewire metabolism to favour the generation of specialized metabolites that support tumour growth and reshape the tumour microenvironment1,2. Lysine functions as a biosynthetic molecule, energy source and antioxidant3-5, but little is known about its pathological role in cancer. Here we show that glioblastoma stem cells (GSCs) reprogram lysine catabolism through the upregulation of lysine transporter SLC7A2 and crotonyl-coenzyme A (crotonyl-CoA)-producing enzyme glutaryl-CoA dehydrogenase (GCDH) with downregulation of the crotonyl-CoA hydratase enoyl-CoA hydratase short chain 1 (ECHS1), leading to accumulation of intracellular crotonyl-CoA and histone H4 lysine crotonylation. A reduction in histone lysine crotonylation by either genetic manipulation or lysine restriction impaired tumour growth. In the nucleus, GCDH interacts with the crotonyltransferase CBP to promote histone lysine crotonylation. Loss of histone lysine crotonylation promotes immunogenic cytosolic double-stranded RNA (dsRNA) and dsDNA generation through enhanced H3K27ac, which stimulates the RNA sensor MDA5 and DNA sensor cyclic GMP-AMP synthase (cGAS) to boost type I interferon signalling, leading to compromised GSC tumorigenic potential and elevated CD8+ T cell infiltration. A lysine-restricted diet synergized with MYC inhibition or anti-PD-1 therapy to slow tumour growth. Collectively, GSCs co-opt lysine uptake and degradation to shunt the production of crotonyl-CoA, remodelling the chromatin landscape to evade interferon-induced intrinsic effects on GSC maintenance and extrinsic effects on immune response.


Subject(s)
Histones , Lysine , Neoplasms , Protein Processing, Post-Translational , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Histones/chemistry , Histones/metabolism , Lysine/deficiency , Lysine/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , RNA, Double-Stranded/immunology , Humans , Animals , Mice , Interferon Type I/immunology
3.
Cell ; 149(6): 1269-83, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682249

ABSTRACT

Cell-cycle arrest, apoptosis, and senescence are widely accepted as the major mechanisms by which p53 inhibits tumor formation. Nevertheless, it remains unclear whether they are the rate-limiting steps in tumor suppression. Here, we have generated mice bearing lysine to arginine mutations at one (p53(K117R)) or three (p53(3KR); K117R+K161R+K162R) of p53 acetylation sites. Although p53(K117R/K117R) cells are competent for p53-mediated cell-cycle arrest and senescence, but not apoptosis, all three of these processes are ablated in p53(3KR/3KR) cells. Surprisingly, unlike p53 null mice, which rapidly succumb to spontaneous thymic lymphomas, early-onset tumor formation does not occur in either p53(K117R/K117R) or p53(3KR/3KR) animals. Notably, p53(3KR) retains the ability to regulate energy metabolism and reactive oxygen species production. These findings underscore the crucial role of acetylation in differentially modulating p53 responses and suggest that unconventional activities of p53, such as metabolic regulation and antioxidant function, are critical for suppression of early-onset spontaneous tumorigenesis.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , Cellular Senescence , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Fibroblasts/metabolism , Gene Knock-In Techniques , Humans , Lymphoma/metabolism , Mice , Molecular Sequence Data , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Sequence Alignment , Thymus Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics
4.
Cell ; 150(3): 620-32, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863012

ABSTRACT

Brown adipose tissue (BAT) can disperse stored energy as heat. Promoting BAT-like features in white adipose (WAT) is an attractive, if elusive, therapeutic approach to staunch the current obesity epidemic. Here we report that gain of function of the NAD-dependent deacetylase SirT1 or loss of function of its endogenous inhibitor Deleted in breast cancer-1 (Dbc1) promote "browning" of WAT by deacetylating peroxisome proliferator-activated receptor (Ppar)-γ on Lys268 and Lys293. SirT1-dependent deacetylation of Lys268 and Lys293 is required to recruit the BAT program coactivator Prdm16 to Pparγ, leading to selective induction of BAT genes and repression of visceral WAT genes associated with insulin resistance. An acetylation-defective Pparγ mutant induces a brown phenotype in white adipocytes, whereas an acetylated mimetic fails to induce "brown" genes but retains the ability to activate "white" genes. We propose that SirT1-dependent Pparγ deacetylation is a form of selective Pparγ modulation of potential therapeutic import.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , PPAR gamma/metabolism , Sirtuin 1/metabolism , 3T3 Cells , Acetylation , Adult , Amino Acid Sequence , Animals , Cells, Cultured , Energy Metabolism , Female , Humans , Insulin Resistance , Ligands , Lysine/analysis , Lysine/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Obesity/complications , Obesity/metabolism , PPAR gamma/chemistry , Resveratrol , Sequence Alignment , Sirtuin 1/chemistry , Sirtuin 1/genetics , Stilbenes/pharmacology , Thermogenesis , Thiazolidinediones/pharmacology
5.
Cell ; 146(6): 1016-28, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925322

ABSTRACT

We report the identification of 67 previously undescribed histone modifications, increasing the current number of known histone marks by about 70%. We further investigated one of the marks, lysine crotonylation (Kcr), confirming that it represents an evolutionarily-conserved histone posttranslational modification. The unique structure and genomic localization of histone Kcr suggest that it is mechanistically and functionally different from histone lysine acetylation (Kac). Specifically, in both human somatic and mouse male germ cell genomes, histone Kcr marks either active promoters or potential enhancers. In male germinal cells immediately following meiosis, Kcr is enriched on sex chromosomes and specifically marks testis-specific genes, including a significant proportion of X-linked genes that escape sex chromosome inactivation in haploid cells. These results therefore dramatically extend the repertoire of histone PTM sites and designate Kcr as a specific mark of active sex chromosome-linked genes in postmeiotic male germ cells.


Subject(s)
Gene Expression Regulation , Histone Code , Animals , HeLa Cells , Histones/chemistry , Histones/metabolism , Humans , Lysine/metabolism , Male , Meiosis , Mice , Protein Processing, Post-Translational , Testis/cytology , Testis/metabolism
6.
Cell ; 146(6): 969-79, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21906795

ABSTRACT

Acetylation of histone and nonhistone proteins is an important posttranslational modification affecting many cellular processes. Here, we report that NuA4 acetylation of Sip2, a regulatory ß subunit of the Snf1 complex (yeast AMP-activated protein kinase), decreases as cells age. Sip2 acetylation, controlled by antagonizing NuA4 acetyltransferase and Rpd3 deacetylase, enhances interaction with Snf1, the catalytic subunit of Snf1 complex. Sip2-Snf1 interaction inhibits Snf1 activity, thus decreasing phosphorylation of a downstream target, Sch9 (homolog of Akt/S6K), and ultimately leading to slower growth but extended replicative life span. Sip2 acetylation mimetics are more resistant to oxidative stress. We further demonstrate that the anti-aging effect of Sip2 acetylation is independent of extrinsic nutrient availability and TORC1 activity. We propose a protein acetylation-phosphorylation cascade that regulates Sch9 activity, controls intrinsic aging, and extends replicative life span in yeast.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Trans-Activators/metabolism , Acetylation , Caloric Restriction , Cell Division , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Protein Kinases/metabolism , Saccharomyces cerevisiae/enzymology , Transcription Factors/metabolism
7.
Mol Cell ; 70(4): 663-678.e6, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29775581

ABSTRACT

Lysine 2-hydroxyisobutyrylation (Khib) is an evolutionarily conserved and widespread histone mark like lysine acetylation (Kac). Here we report that p300 functions as a lysine 2-hyroxyisobutyryltransferase to regulate glycolysis in response to nutritional cues. We discovered that p300 differentially regulates Khib and Kac on distinct lysine sites, with only 6 of the 149 p300-targeted Khib sites overlapping with the 693 p300-targeted Kac sites. We demonstrate that diverse cellular proteins, particularly glycolytic enzymes, are targeted by p300 for Khib, but not for Kac. Specifically, deletion of p300 significantly reduces Khib levels on several p300-dependent, Khib-specific sites on key glycolytic enzymes including ENO1, decreasing their catalytic activities. Consequently, p300-deficient cells have impaired glycolysis and are hypersensitive to glucose-depletion-induced cell death. Our study reveals an p300-catalyzed, Khib-specific molecular mechanism that regulates cellular glucose metabolism and further indicate that p300 has an intrinsic ability to select short-chain acyl-CoA-dependent protein substrates.


Subject(s)
E1A-Associated p300 Protein/metabolism , Glucose/metabolism , Glycolysis , Histones/metabolism , Hydroxybutyrates/metabolism , Lysine/metabolism , Proteome/metabolism , Acetylation , E1A-Associated p300 Protein/genetics , Histones/genetics , Humans , Lysine/genetics
8.
Nature ; 574(7779): 575-580, 2019 10.
Article in English | MEDLINE | ID: mdl-31645732

ABSTRACT

The Warburg effect, which originally described increased production of lactate in cancer, is associated with diverse cellular processes such as angiogenesis, hypoxia, polarization of macrophages and activation of T cells. This phenomenon is intimately linked to several diseases including neoplasia, sepsis and autoimmune diseases1,2. Lactate, which is converted from pyruvate in tumour cells, is widely known as an energy source and metabolic by-product. However, its non-metabolic functions in physiology and disease remain unknown. Here we show that lactate-derived lactylation of histone lysine residues serves as an epigenetic modification that directly stimulates gene transcription from chromatin. We identify 28 lactylation sites on core histones in human and mouse cells. Hypoxia and bacterial challenges induce the production of lactate by glycolysis, and this acts as a precursor that stimulates histone lactylation. Using M1 macrophages that have been exposed to bacteria as a model system, we show that histone lactylation has different temporal dynamics from acetylation. In the late phase of M1 macrophage polarization, increased histone lactylation induces homeostatic genes that are involved in wound healing, including Arg1. Collectively, our results suggest that an endogenous 'lactate clock' in bacterially challenged M1 macrophages turns on gene expression to promote homeostasis. Histone lactylation thus represents an opportunity to improve our understanding of the functions of lactate and its role in diverse pathophysiological conditions, including infection and cancer.


Subject(s)
Epigenesis, Genetic , Glycolysis/genetics , Histones/chemistry , Histones/metabolism , Lactic Acid/metabolism , Acetylation , Amino Acid Sequence , Animals , Cell Line, Tumor , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Homeostasis , Humans , Hypoxia/metabolism , Lysine/chemistry , Lysine/metabolism , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results , Transcription, Genetic
10.
Cell ; 139(7): 1290-302, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064375

ABSTRACT

Polycomb Repressive Complex 2 (PRC2) regulates key developmental genes in embryonic stem (ES) cells and during development. Here we show that Jarid2/Jumonji, a protein enriched in pluripotent cells and a founding member of the Jumonji C (JmjC) domain protein family, is a PRC2 subunit in ES cells. Genome-wide ChIP-seq analyses of Jarid2, Ezh2, and Suz12 binding reveal that Jarid2 and PRC2 occupy the same genomic regions. We further show that Jarid2 promotes PRC2 recruitment to the target genes while inhibiting PRC2 histone methyltransferase activity, suggesting that it acts as a "molecular rheostat" that finely calibrates PRC2 functions at developmental genes. Using Xenopus laevis as a model we demonstrate that Jarid2 knockdown impairs the induction of gastrulation genes in blastula embryos and results in failure of differentiation. Our findings illuminate a mechanism of histone methylation regulation in pluripotent cells and during early cell-fate transitions.


Subject(s)
Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , Humans , Mice , Mitochondria/metabolism , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , RNA/metabolism , Retinoblastoma-Binding Protein 2/metabolism
11.
Cell ; 136(6): 1073-84, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19303850

ABSTRACT

Histone acetyltransferases (HATs) and histone deacetylases (HDACs) conduct many critical functions through nonhistone substrates in metazoans, but only chromatin-associated nonhistone substrates are known in Saccharomyces cerevisiae. Using yeast proteome microarrays, we identified and validated many nonchromatin substrates of the essential nucleosome acetyltransferase of H4 (NuA4) complex. Among these, acetylation sites (Lys19 and 514) of phosphoenolpyruvate carboxykinase (Pck1p) were determined by tandem mass spectrometry. Acetylation at Lys514 was crucial for enzymatic activity and the ability of yeast cells to grow on nonfermentable carbon sources. Furthermore, Sir2p deacetylated Pck1p both in vitro and in vivo. Loss of Pck1p activity blocked the extension of yeast chronological life span caused by water starvation. In human hepatocellular carcinoma (HepG2) cells, human Pck1 acetylation and glucose production were dependent on TIP60, the human homolog of ESA1. Our findings demonstrate a regulatory function for the NuA4 complex in glucose metabolism and life span by acetylating a critical metabolic enzyme.


Subject(s)
Gluconeogenesis , Histone Acetyltransferases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Acetylation , Gene Knockdown Techniques , Glucose/metabolism , Histone Acetyltransferases/genetics , Histone Deacetylases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysine Acetyltransferase 5 , Multiprotein Complexes/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Protein Array Analysis , Sirtuins/metabolism , Water/metabolism
12.
Mol Cell ; 62(2): 181-193, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27105114

ABSTRACT

Recognition of histone covalent modifications by chromatin-binding protein modules ("readers") constitutes a major mechanism for epigenetic regulation, typified by bromodomains that bind acetyllysine. Non-acetyl histone lysine acylations (e.g., crotonylation, butyrylation, propionylation) have been recently identified, but readers that prefer these acylations have not been characterized. Here we report that the AF9 YEATS domain displays selectively higher binding affinity for crotonyllysine over acetyllysine. Structural studies revealed an extended aromatic sandwiching cage with crotonyl specificity arising from π-aromatic and hydrophobic interactions between crotonyl and aromatic rings. These features are conserved among the YEATS, but not the bromodomains. Using a cell-based model, we showed that AF9 co-localizes with crotonylated histone H3 and positively regulates gene expression in a YEATS domain-dependent manner. Our studies define the evolutionarily conserved YEATS domain as a family of crotonyllysine readers and specifically demonstrate that the YEATS domain of AF9 directly links histone crotonylation to active transcription.


Subject(s)
Crotonates/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Transcription, Genetic , Transcriptional Activation , Acetylation , Animals , Binding Sites , Chromatin Assembly and Disassembly , Epigenesis, Genetic , HEK293 Cells , Histones/chemistry , Histones/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Lysine , Mice , Models, Molecular , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Domains , RAW 264.7 Cells , RNA-Binding Proteins/metabolism , Transcription Factors , Transfection
13.
Mol Cell ; 62(2): 169-180, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27105113

ABSTRACT

Recently discovered histone lysine acylation marks increase the functional diversity of nucleosomes well beyond acetylation. Here, we focus on histone butyrylation in the context of sperm cell differentiation. Specifically, we investigate the butyrylation of histone H4 lysine 5 and 8 at gene promoters where acetylation guides the binding of Brdt, a bromodomain-containing protein, thereby mediating stage-specific gene expression programs and post-meiotic chromatin reorganization. Genome-wide mapping data show that highly active Brdt-bound gene promoters systematically harbor competing histone acetylation and butyrylation marks at H4 K5 and H4 K8. Despite acting as a direct stimulator of transcription, histone butyrylation competes with acetylation, especially at H4 K5, to prevent Brdt binding. Additionally, H4 K5K8 butyrylation also marks retarded histone removal during late spermatogenesis. Hence, alternating H4 acetylation and butyrylation, while sustaining direct gene activation and dynamic bromodomain binding, could impact the final male epigenome features.


Subject(s)
Butyrates/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histones/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Processing, Post-Translational , Spermatocytes/metabolism , Acetylation , Animals , Binding Sites , Cell Differentiation , Chromatin Assembly and Disassembly , Genome-Wide Association Study , Histones/chemistry , Histones/genetics , Lysine , Male , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Conformation , Structure-Activity Relationship , Transcription, Genetic , Transcriptional Activation
14.
Mol Cell ; 61(2): 199-209, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26725009

ABSTRACT

Mitochondrial metabolism is necessary for the maintenance of oxidative TCA cycle function and mitochondrial membrane potential. Previous attempts to decipher whether mitochondria are necessary for biological outcomes have been hampered by genetic and pharmacologic methods that simultaneously disrupt multiple functions linked to mitochondrial metabolism. Here, we report that inducible depletion of mitochondrial DNA (ρ(ο) cells) diminished respiration, oxidative TCA cycle function, and the mitochondrial membrane potential, resulting in diminished cell proliferation, hypoxic activation of HIF-1, and specific histone acetylation marks. Genetic reconstitution only of the oxidative TCA cycle function specifically in these inducible ρ(ο) cells restored metabolites, resulting in re-establishment of histone acetylation. In contrast, genetic reconstitution of the mitochondrial membrane potential restored ROS, which were necessary for hypoxic activation of HIF-1 and cell proliferation. These results indicate that distinct mitochondrial functions associated with respiration are necessary for cell proliferation, epigenetics, and HIF-1 activation.


Subject(s)
Citric Acid Cycle , Membrane Potential, Mitochondrial , Acetylation , Cell Proliferation , Cell Respiration , DNA Polymerase gamma , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/metabolism , HEK293 Cells , Histones/metabolism , Humans , Hypoxia-Inducible Factor 1/metabolism , Metabolome , Mitochondrial Proteins/metabolism , Oxidation-Reduction , Oxidoreductases/metabolism , Oxygen Consumption , Plant Proteins/metabolism , Protein Stability , Reactive Oxygen Species/metabolism
15.
Mol Cell ; 62(2): 194-206, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27105115

ABSTRACT

Here we report the identification and verification of a ß-hydroxybutyrate-derived protein modification, lysine ß-hydroxybutyrylation (Kbhb), as a new type of histone mark. Histone Kbhb marks are dramatically induced in response to elevated ß-hydroxybutyrate levels in cultured cells and in livers from mice subjected to prolonged fasting or streptozotocin-induced diabetic ketoacidosis. In total, we identified 44 histone Kbhb sites, a figure comparable to the known number of histone acetylation sites. By ChIP-seq and RNA-seq analysis, we demonstrate that histone Kbhb is a mark enriched in active gene promoters and that the increased H3K9bhb levels that occur during starvation are associated with genes upregulated in starvation-responsive metabolic pathways. Histone ß-hydroxybutyrylation thus represents a new epigenetic regulatory mark that couples metabolism to gene expression, offering a new avenue to study chromatin regulation and diverse functions of ß-hydroxybutyrate in the context of important human pathophysiological states, including diabetes, epilepsy, and neoplasia.


Subject(s)
Diabetic Ketoacidosis/metabolism , Energy Metabolism , Gene Expression Regulation , Histones/metabolism , Hydroxybutyrates/metabolism , Liver/metabolism , Protein Processing, Post-Translational , Starvation/metabolism , Animals , Binding Sites , Chromatin Assembly and Disassembly , Diabetic Ketoacidosis/chemically induced , Diabetic Ketoacidosis/genetics , Disease Models, Animal , Epigenesis, Genetic , Fatty Acids/metabolism , Glucose/metabolism , HEK293 Cells , Histones/genetics , Humans , Lysine , Mice, Inbred C57BL , Promoter Regions, Genetic , Starvation/genetics , Streptozocin
16.
Sensors (Basel) ; 24(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38931787

ABSTRACT

Grating-type spectral imaging systems are frequently employed in scenes for high-resolution remote-sensing observations of the Earth. However, the entrance of the grating-type spectral imaging system is a slit or a pinhole. This structure relies on the push broom method, which presents a challenge in capturing spectral information of transiently changing targets. To address this issue, the IFU is used to slice the focal plane of the telescope system, thereby expanding the instantaneous field of view (IFOV) of the grating-type spectral imaging system. The aberrations introduced by the expansion of the single-slice field of view (FOV) of the IFU are corrected, and the conversion of the IFU's FOV from arcseconds to degrees is achieved. The design of a spectral imaging system based on an image-slicer IFU for remote sensing is finally completed. The system has a wavelength range of 1400 nm to 2000 nm, and a spectral resolution of better than 3 nm. Compared with the traditional grating-type spectral imaging system, its IFOV is expanded by a factor of four. And it allows for the capture of complete spectral information of transiently changing targets through a single exposure. The simulation results demonstrate that the system has good performance at each sub-slit, thereby validating the effectiveness and advantages of the proposed system for dynamic target capture in remote sensing.

17.
Opt Express ; 31(26): 43198-43212, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38178419

ABSTRACT

After entering the 21st century, asteroid exploration has become one of the key research directions in the field of deep space exploration. In this article, we present a navigation, imaging, and hyperspectral acquisition integrated optical system designed for asteroid exploration. Based on the pixel-level light modulation capability of the digital micromirror device (DMD), this system introduces a triple-pass total internal reflection (TIR) prism to overcome the limitation of small DMD deflection angles, achieving the co-aperture design of the high-resolution imaging branch and the hyperspectral acquisition branch. In view of the actual usage scenarios, we design a new triple-pass TIR prism and optimize it, correcting the on-axis point coma and astigmatism it introduces, and designing it to be lightweight and compact. Compared to existing optical systems for asteroid exploration, this system offers advantages such as compact structure, no moving parts, high spatial resolution, high spectral resolution, and more flexible imaging modes.

18.
Cell ; 133(4): 612-26, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18485870

ABSTRACT

The activation of the tumor suppressor p53 facilitates the cellular response to genotoxic stress; however, the p53 response can only be executed if its interaction with its inhibitor Mdm2 is abolished. There have been conflicting reports on the question of whether p53 posttranslational modifications, such as phosphorylation or acetylation, are essential or only play a subtle, fine-tuning role in the p53 response. Thus, it remains unclear whether p53 modification is absolutely required for its activation. We have now identified all major acetylation sites of p53. Although unacetylated p53 retains its ability to induce the p53-Mdm2 feedback loop, loss of acetylation completely abolishes p53-dependent growth arrest and apoptosis. Notably, acetylation of p53 abrogates Mdm2-mediated repression by blocking the recruitment of Mdm2 to p53-responsive promoters, which leads to p53 activation independent of its phosphorylation status. Our study identifies p53 acetylation as an indispensable event that destabilizes the p53-Mdm2 interaction and enables the p53-mediated stress response.


Subject(s)
Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Acetylation , Amino Acid Sequence , Animals , Apoptosis , CREB-Binding Protein/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Sequence Alignment , Tandem Mass Spectrometry , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Ubiquitination
19.
Mol Cell ; 58(2): 203-15, 2015 04 16.
Article in English | MEDLINE | ID: mdl-25818647

ABSTRACT

Acetylation of histones at DNA regulatory elements plays a critical role in transcriptional activation. Histones are also modified by other acyl moieties, including crotonyl, yet the mechanisms that govern acetylation versus crotonylation and the functional consequences of this "choice" remain unclear. We show that the coactivator p300 has both crotonyltransferase and acetyltransferase activities, and that p300-catalyzed histone crotonylation directly stimulates transcription to a greater degree than histone acetylation. Levels of histone crotonylation are regulated by the cellular concentration of crotonyl-CoA, which can be altered through genetic and environmental perturbations. In a cell-based model of transcriptional activation, increasing or decreasing the cellular concentration of crotonyl-CoA leads to enhanced or diminished gene expression, respectively, which correlates with the levels of histone crotonylation flanking the regulatory elements of activated genes. Our findings support a general principle wherein differential histone acylation (i.e., acetylation versus crotonylation) couples cellular metabolism to the regulation of gene expression.


Subject(s)
Acyl Coenzyme A/metabolism , E1A-Associated p300 Protein/metabolism , Histones/metabolism , Macrophages/immunology , RNA, Messenger/metabolism , Transcriptional Activation , Acetate-CoA Ligase/genetics , Acetate-CoA Ligase/metabolism , Acetylation , Acyl Coenzyme A/genetics , Cell Line , Cell-Free System , E1A-Associated p300 Protein/genetics , HEK293 Cells , HeLa Cells , Humans , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Molecular Sequence Data
20.
Nucleic Acids Res ; 49(1): 177-189, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33313896

ABSTRACT

Short-chain acylations of lysine residues in eukaryotic proteins are recognized as essential posttranslational chemical modifications (PTMs) that regulate cellular processes from transcription, cell cycle, metabolism, to signal transduction. Lysine butyrylation was initially discovered as a normal straight chain butyrylation (Knbu). Here we report its structural isomer, branched chain butyrylation, i.e. lysine isobutyrylation (Kibu), existing as a new PTM on nuclear histones. Uniquely, isobutyryl-CoA is derived from valine catabolism and branched chain fatty acid oxidation which is distinct from the metabolism of n-butyryl-CoA. Several histone acetyltransferases were found to possess lysine isobutyryltransferase activity in vitro, especially p300 and HAT1. Transfection and western blot experiments showed that p300 regulated histone isobutyrylation levels in the cell. We resolved the X-ray crystal structures of HAT1 in complex with isobutyryl-CoA that gleaned an atomic level insight into HAT-catalyzed isobutyrylation. RNA-Seq profiling revealed that isobutyrate greatly affected the expression of genes associated with many pivotal biological pathways. Together, our findings identify Kibu as a novel chemical modification mark in histones and suggest its extensive role in regulating epigenetics and cellular physiology.


Subject(s)
Histone Code , Isobutyrates/metabolism , Lysine Acetyltransferases/metabolism , Protein Processing, Post-Translational , Acyl Coenzyme A/chemical synthesis , Acyl Coenzyme A/metabolism , Acylation , Amino Acid Sequence , Chromatography, High Pressure Liquid , Crystallography, X-Ray , HEK293 Cells , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Isobutyrates/pharmacology , Models, Molecular , Protein Conformation , Protein Domains , Recombinant Proteins/metabolism , Tandem Mass Spectrometry , Valine/metabolism , p300-CBP Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL