Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.171
Filter
1.
PLoS Pathog ; 20(6): e1012238, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38843141

ABSTRACT

Although lack of ADAR (adenosine deaminase acting on RNA) orthologs, genome-wide A-to-I editing occurs specifically during sexual reproduction in a number of filamentous ascomycetes, including Fusarium graminearum and Neurospora crassa. Unlike ADAR-mediated editing in animals, fungal A-to-I editing has a strong preference for hairpin loops and U at -1 position, which leads to frequent editing of UAG and UAA stop codons. Majority of RNA editing events in fungi are in the coding region and cause amino acid changes. Some of these editing events have been experimentally characterized for providing heterozygote and adaptive advantages in F. graminearum. Recent studies showed that FgTad2 and FgTad3, 2 ADAT (adenosine deaminase acting on tRNA) enzymes that normally catalyze the editing of A34 in the anticodon of tRNA during vegetative growth mediate A-to-I mRNA editing during sexual reproduction. Stage specificity of RNA editing is conferred by stage-specific expression of short transcript isoforms of FgTAD2 and FgTAD3 as well as cofactors such as AME1 and FIP5 that facilitate the editing of mRNA in perithecia. Taken together, fungal A-to-I RNA editing during sexual reproduction is catalyzed by ADATs and it has the same sequence and structural preferences with editing of A34 in tRNA.


Subject(s)
Adenosine Deaminase , RNA Editing , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Ascomycota/genetics , RNA, Fungal/genetics , RNA, Fungal/metabolism , Adenosine/metabolism , Adenosine/genetics , Inosine/metabolism , Inosine/genetics , Fusarium/genetics , Neurospora crassa/genetics
2.
BMC Biol ; 22(1): 106, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715001

ABSTRACT

BACKGROUND: The significance of A-to-I RNA editing in nervous system development is widely recognized; however, its influence on retina development remains to be thoroughly understood. RESULTS: In this study, we performed RNA sequencing and ribosome profiling experiments on developing mouse retinas to characterize the temporal landscape of A-to-I editing. Our findings revealed temporal changes in A-to-I editing, with distinct editing patterns observed across different developmental stages. Further analysis showed the interplay between A-to-I editing and alternative splicing, with A-to-I editing influencing splicing efficiency and the quantity of splicing events. A-to-I editing held the potential to enhance translation diversity, but this came at the expense of reduced translational efficiency. When coupled with splicing, it could produce a coordinated effect on gene translation. CONCLUSIONS: Overall, this study presents a temporally resolved atlas of A-to-I editing, connecting its changes with the impact on alternative splicing and gene translation in retina development.


Subject(s)
Protein Biosynthesis , RNA Editing , Retina , Animals , Mice , Retina/metabolism , Retina/embryology , Alternative Splicing , Inosine/metabolism , Inosine/genetics , Adenosine/metabolism
3.
BMC Genomics ; 25(1): 431, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693480

ABSTRACT

Ophthalmic manifestations have recently been observed in acute and post-acute complications of COVID-19 caused by SARS-CoV-2 infection. Our precious study has shown that host RNA editing is linked to RNA viral infection, yet ocular adenosine to inosine (A-to-I) RNA editing during SARS-CoV-2 infection remains uninvestigated in COVID-19. Herein we used an epitranscriptomic pipeline to analyze 37 samples and investigate A-to-I editing associated with SARS-CoV-2 infection, in five ocular tissue types including the conjunctiva, limbus, cornea, sclera, and retinal organoids. Our results revealed dramatically altered A-to-I RNA editing across the five ocular tissues. Notably, the transcriptome-wide average level of RNA editing was increased in the cornea but generally decreased in the other four ocular tissues. Functional enrichment analysis showed that differential RNA editing (DRE) was mainly in genes related to ubiquitin-dependent protein catabolic process, transcriptional regulation, and RNA splicing. In addition to tissue-specific RNA editing found in each tissue, common RNA editing was observed across different tissues, especially in the innate antiviral immune gene MAVS and the E3 ubiquitin-protein ligase MDM2. Analysis in retinal organoids further revealed highly dynamic RNA editing alterations over time during SARS-CoV-2 infection. Our study thus suggested the potential role played by RNA editing in ophthalmic manifestations of COVID-19, and highlighted its potential transcriptome impact, especially on innate immunity.


Subject(s)
COVID-19 , RNA Editing , SARS-CoV-2 , Humans , COVID-19/genetics , COVID-19/virology , SARS-CoV-2/genetics , Adenosine/metabolism , Inosine/metabolism , Inosine/genetics , Transcriptome , Eye/metabolism , Eye/virology
4.
Food Res Int ; 183: 114208, 2024 May.
Article in English | MEDLINE | ID: mdl-38760138

ABSTRACT

To explore the underlying mechanisms by which superchilling (SC, -3 °C within 5 h of slaughter) improves beef tenderness, an untargeted metabolomics strategy was employed. M. Longissimus lumborum (LL) muscles from twelve beef carcasses were assigned to either SC or very fast chilling (VFC, 0 °C within 5 h of slaughter) treatments, with conventional chilling (CC, 0 âˆ¼ 4 °C until 24 h post-mortem) serving as the control (6 per group). Biochemical properties and metabolites were investigated during the early post-mortem period. The results showed that the degradation of µ-calpain and caspase 3 occurred earlier in SC treated sample, which might be attributed to the accelerated accumulation of free Ca2+. The metabolomic profiles of samples from the SC and CC treatments were clearly distinguished based on partial least squares-discriminant analysis (PLS-DA) at each time point. It is noteworthy that more IMP and 4-hydroxyproline were found in the comparison between SC and CC treatments. According to the results of metabolic pathways analysis and the correlation analysis between traits related to tenderness and metabolites with significant differences (SC vs. CC), it can be suggested that the tenderization effect of the SC treatment may be related to the alteration of arginine and proline metabolism, and purine metabolism in the early post-mortem phase.


Subject(s)
Metabolomics , Muscle, Skeletal , Red Meat , Animals , Metabolomics/methods , Cattle , Red Meat/analysis , Muscle, Skeletal/metabolism , Muscle, Skeletal/chemistry , Cold Temperature , Food Handling/methods , Chromatography, Liquid , Caspase 3/metabolism , Discriminant Analysis , Postmortem Changes , Calpain/metabolism , Least-Squares Analysis , Proline/metabolism , Mass Spectrometry/methods , Inosine/metabolism , Inosine/analysis , Liquid Chromatography-Mass Spectrometry
5.
Nat Commun ; 15(1): 4049, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744925

ABSTRACT

Nanopore direct RNA sequencing (DRS) has emerged as a powerful tool for RNA modification identification. However, concurrently detecting multiple types of modifications in a single DRS sample remains a challenge. Here, we develop TandemMod, a transferable deep learning framework capable of detecting multiple types of RNA modifications in single DRS data. To train high-performance TandemMod models, we generate in vitro epitranscriptome datasets from cDNA libraries, containing thousands of transcripts labeled with various types of RNA modifications. We validate the performance of TandemMod on both in vitro transcripts and in vivo human cell lines, confirming its high accuracy for profiling m6A and m5C modification sites. Furthermore, we perform transfer learning for identifying other modifications such as m7G, Ψ, and inosine, significantly reducing training data size and running time without compromising performance. Finally, we apply TandemMod to identify 3 types of RNA modifications in rice grown in different environments, demonstrating its applicability across species and conditions. In summary, we provide a resource with ground-truth labels that can serve as benchmark datasets for nanopore-based modification identification methods, and TandemMod for identifying diverse RNA modifications using a single DRS sample.


Subject(s)
Oryza , Sequence Analysis, RNA , Humans , Sequence Analysis, RNA/methods , Oryza/genetics , RNA Processing, Post-Transcriptional , Nanopores , RNA/genetics , RNA/metabolism , Nanopore Sequencing/methods , Deep Learning , Inosine/metabolism , Inosine/genetics , Transcriptome/genetics
6.
Nat Commun ; 15(1): 3934, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729938

ABSTRACT

A-to-I mRNA editing in animals is mediated by ADARs, but the mechanism underlying sexual stage-specific A-to-I mRNA editing in fungi remains unknown. Here, we show that the eukaryotic tRNA-specific heterodimeric deaminase FgTad2-FgTad3 is responsible for A-to-I mRNA editing in Fusarium graminearum. This editing capacity relies on the interaction between FgTad3 and a sexual stage-specific protein called Ame1. Although Ame1 orthologs are widely distributed in fungi, the interaction originates in Sordariomycetes. We have identified key residues responsible for the FgTad3-Ame1 interaction. The expression and activity of FgTad2-FgTad3 are regulated through alternative promoters, alternative translation initiation, and post-translational modifications. Our study demonstrates that the FgTad2-FgTad3-Ame1 complex can efficiently edit mRNA in yeasts, bacteria, and human cells, with important implications for the development of base editors in therapy and agriculture. Overall, this study uncovers mechanisms, regulation, and evolution of RNA editing in fungi, highlighting the role of protein-protein interactions in modulating deaminase function.


Subject(s)
Fungal Proteins , Fusarium , RNA Editing , RNA, Messenger , Fusarium/genetics , Fusarium/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Humans , Gene Expression Regulation, Fungal , Evolution, Molecular , Protein Processing, Post-Translational , Inosine/metabolism , Inosine/genetics
7.
Anal Chem ; 96(21): 8730-8739, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38743814

ABSTRACT

Adenosine-to-inosine (A-to-I) editing and N6-methyladenosine (m6A) modifications are pivotal RNA modifications with widespread functional significance in physiological and pathological processes. Although significant effort has been dedicated to developing methodologies for identifying and quantifying these modifications, traditional approaches have often focused on each modification independently, neglecting the potential co-occurrence of A-to-I editing and m6A modifications at the same adenosine residues. This limitation has constrained our understanding of the intricate regulatory mechanisms governing RNA function and the interplay between different types of RNA modifications. To address this gap, we introduced an innovative technique called deamination-assisted reverse transcription stalling (DARTS), specifically designed for the simultaneous quantification of A-to-I editing and m6A at the same RNA sites. DARTS leverages the selective deamination activity of the engineered TadA-TadA8e protein, which converts adenosine residues to inosine, in combination with the unique property of Bst 2.0 DNA polymerase, which stalls when encountering inosine during reverse transcription. This approach enables the accurate quantification of A-to-I editing, m6A, and unmodified adenosine at identical RNA sites. The DARTS method is remarkable for its ability to directly quantify two distinct types of RNA modifications simultaneously, a capability that has remained largely unexplored in the field of RNA biology. By facilitating a comprehensive analysis of the co-occurrence and interaction between A-to-I editing and m6A modifications, DARTS opens new avenues for exploring the complex regulatory networks modulated by different RNA modifications.


Subject(s)
Adenosine , Inosine , RNA Editing , Adenosine/analogs & derivatives , Adenosine/analysis , Adenosine/metabolism , Inosine/metabolism , Inosine/analogs & derivatives , Inosine/chemistry , Deamination , RNA/metabolism , RNA/genetics , RNA/analysis , Reverse Transcription , Humans
9.
Mol Genet Genomics ; 299(1): 46, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38642133

ABSTRACT

Adenosine-to-inosine (A-to-I) RNA editing, resembling A-to-G mutation, confers adaptiveness by increasing proteomic diversity in a temporal-spatial manner. This evolutionary theory named "proteomic diversifying hypothesis" has only partially been tested in very few organisms like Drosophila melanogaster, mainly by observing the positive selection on nonsynonymous editing events. To find additional genome-wide evidences supporting this interesting assumption, we retrieved the genomes of four Drosophila species and collected 20 deep-sequenced transcriptomes of different developmental stages and neuron populations of D. melanogaster. We systematically profiled the RNA editomes in these samples and performed meticulous comparative genomic analyses. Further evidences were found to support the diversifying hypothesis. (1) None of the nonsynonymous editing sites in D. melanogaster had ancestral G-alleles, while the silent editing sites had an unignorable fraction of ancestral G-alleles; (2) Only very few nonsynonymous editing sites in D. melanogaster had corresponding G-alleles derived in the genomes of sibling species, and the fraction of such situation was significantly lower than that of silent editing sites; (3) The few nonsynonymous editing with corresponding G-alleles had significantly more variable editing levels (across samples) than other nonsynonymous editing sites in D. melanogaster. The proteomic diversifying nature of RNA editing in Drosophila excludes the restorative role which favors an ancestral G-allele. The few fixed G-alleles in sibling species might facilitate the adaptation to particular environment and the corresponding nonsynonymous editing in D. melanogaster would introduce stronger advantage of flexible proteomic diversification. With multi-Omics data, our study consolidates the nature of evolutionary significance of A-to-I RNA editing sites in model insects.


Subject(s)
Drosophila melanogaster , RNA , Animals , RNA/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Proteomics , RNA Editing/genetics , Adenosine/genetics , Adenosine/metabolism , Inosine/genetics , Inosine/metabolism , Genomics , Drosophila/genetics
10.
RNA ; 30(5): 512-520, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38531652

ABSTRACT

Inosine (I), resulting from the deamination of adenosine (A), is a prominent modification in the human transcriptome. The enzymes responsible for the conversion of adenosine to inosine in human mRNAs are the ADARs (adenosine deaminases acting on RNA). Inosine modification introduces a layer of complexity to mRNA processing and function, as it can impact various aspects of RNA biology, including mRNA stability, splicing, translation, and protein binding. The relevance of this process is emphasized in the growing number of human disorders associated with dysregulated A-to-I editing pathways. Here, we describe the impact of the A-to-I conversion on the structure and stability of duplex RNA and on the consequences of this modification at different locations in mRNAs. Furthermore, we highlight specific open questions regarding the interplay between inosine formation in duplex RNA and the innate immune response.


Subject(s)
RNA Editing , RNA , Humans , RNA, Messenger/metabolism , RNA/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Inosine/metabolism , Adenosine/genetics , Adenosine/metabolism
11.
RNA ; 30(5): 521-529, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38531651

ABSTRACT

In this article, I recount my memories of key experiments that led to my entry into the RNA editing/modification field. I highlight initial observations made by the pioneers in the ADAR field, and how they fit into our current understanding of this family of enzymes. I discuss early mysteries that have now been solved, as well as those that still linger. Finally, I discuss important, outstanding questions and acknowledge my hope for the future of the RNA editing/modification field.


Subject(s)
Adenosine Deaminase , RNA , RNA/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , RNA Editing , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Inosine/metabolism , RNA, Double-Stranded
12.
Cell Metab ; 36(5): 1013-1029.e5, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38547864

ABSTRACT

Metabolic dysfunction-associated fatty liver disease (MAFLD) has a global prevalence of about 25% and no approved therapy. Using metabolomic and proteomic analyses, we identified high expression of hepatic transketolase (TKT), a metabolic enzyme of the pentose phosphate pathway, in human and mouse MAFLD. Hyperinsulinemia promoted TKT expression through the insulin receptor-CCAAT/enhancer-binding protein alpha axis. Utilizing liver-specific TKT overexpression and knockout mouse models, we demonstrated that TKT was sufficient and required for MAFLD progression. Further metabolic flux analysis revealed that Tkt deletion increased hepatic inosine levels to activate the protein kinase A-cAMP response element binding protein cascade, promote phosphatidylcholine synthesis, and improve mitochondrial function. Moreover, insulin induced hepatic TKT to limit inosine-dependent mitochondrial activity. Importantly, N-acetylgalactosamine (GalNAc)-siRNA conjugates targeting hepatic TKT showed promising therapeutic effects on mouse MAFLD. Our study uncovers how hyperinsulinemia regulates TKT-orchestrated inosine metabolism and mitochondrial function and provides a novel therapeutic strategy for MAFLD prevention and treatment.


Subject(s)
Inosine , Mitochondria , Transketolase , Animals , Female , Humans , Male , Mice , Hyperinsulinism/metabolism , Inosine/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/drug effects , Transketolase/metabolism
13.
FEBS Lett ; 598(9): 1080-1093, 2024 May.
Article in English | MEDLINE | ID: mdl-38523059

ABSTRACT

Recent developments in sequencing and bioinformatics have advanced our understanding of adenosine-to-inosine (A-to-I) RNA editing. Surprisingly, recent analyses have revealed the capability of adenosine deaminase acting on RNA (ADAR) to edit DNA:RNA hybrid strands. However, edited inosines in DNA remain largely unexplored. A precise biochemical method could help uncover these potentially rare DNA editing sites. We explore maleimide as a scaffold for inosine labeling. With fluorophore-conjugated maleimide, we were able to label inosine in RNA or DNA. Moreover, with biotin-conjugated maleimide, we purified RNA and DNA containing inosine. Our novel technique of inosine chemical labeling and affinity molecular purification offers substantial advantages and provides a versatile platform for further discovery of A-to-I editing sites in RNA and DNA.


Subject(s)
Adenosine , Inosine , RNA Editing , Inosine/chemistry , Inosine/metabolism , Adenosine/chemistry , Adenosine/metabolism , Adenosine/analogs & derivatives , Deamination , DNA/chemistry , DNA/metabolism , Maleimides/chemistry , Adenosine Deaminase/metabolism , Adenosine Deaminase/chemistry , RNA/chemistry , RNA/metabolism , Staining and Labeling/methods , Humans , Fluorescent Dyes/chemistry , Biotin/chemistry , Biotin/metabolism
14.
FEBS J ; 291(9): 1992-2008, 2024 May.
Article in English | MEDLINE | ID: mdl-38362806

ABSTRACT

The nucleoside inosine is a main intermediate of purine nucleotide catabolism in Saccharomyces cerevisiae and is produced via the dephosphorylation of inosine monophosphate (IMP) by IMP-specific 5'-nucleotidase 1 (ISN1), which is present in many eukaryotic organisms. Upon transition of yeast from oxidative to fermentative growth, ISN1 is important for intermediate inosine accumulation as purine storage, but details of ISN1 regulation are unknown. We characterized structural and kinetic behavior of ISN1 from S. cerevisiae (ScISN1) and showed that tetrameric ScISN1 is negatively regulated by inosine and adenosine triphosphate (ATP). Regulation involves an inosine-binding allosteric site along with IMP-induced local and global conformational changes in the monomer and a tetrameric re-arrangement, respectively. A proposed interaction network propagates local conformational changes in the active site to the intersubunit interface, modulating the allosteric features of ScISN1. Via ATP and inosine, ScISN1 activity is likely fine-tuned to regulate IMP and inosine homeostasis. These regulatory and catalytic features of ScISN1 contrast with those of the structurally homologous ISN1 from Plasmodium falciparum, indicating that ISN1 enzymes may serve different biological purposes in different organisms.


Subject(s)
Adenosine Triphosphate , Allosteric Site , Inosine , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/metabolism , Inosine/metabolism , Kinetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry , Catalytic Domain , Allosteric Regulation , Crystallography, X-Ray , Inosine Monophosphate/metabolism , Models, Molecular , Protein Conformation , Protein Binding
15.
Science ; 383(6679): 205-211, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38207021

ABSTRACT

Antibodies are produced at high rates to provide immunoprotection, which puts pressure on the B cell translational machinery. Here, we identified a pattern of codon usage conserved across antibody genes. One feature thereof is the hyperutilization of codons that lack genome-encoded Watson-Crick transfer RNAs (tRNAs), instead relying on the posttranscriptional tRNA modification inosine (I34), which expands the decoding capacity of specific tRNAs through wobbling. Antibody-secreting cells had increased I34 levels and were more reliant on I34 for protein production than naïve B cells. Furthermore, antibody I34-dependent codon usage may influence B cell passage through regulatory checkpoints. Our work elucidates the interface between the tRNA pool and protein production in the immune system and has implications for the design and selection of antibodies for vaccines and therapeutics.


Subject(s)
Antibodies , Antibody Formation , B-Lymphocytes , Codon Usage , Immunoglobulin Heavy Chains , Inosine , RNA, Transfer , Antibody Formation/genetics , Codon/genetics , Inosine/genetics , Inosine/metabolism , RNA, Transfer/genetics , Antibodies/genetics , Humans , B-Lymphocytes/immunology , Immunoglobulin Heavy Chains/genetics
16.
Science ; 383(6679): 146-147, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38207031

ABSTRACT

Optimized transfer RNA (tRNA) codon use can speed up antibody generation.


Subject(s)
Antibody Formation , Codon Usage , Inosine , RNA, Transfer , RNA, Transfer/genetics , RNA, Transfer/metabolism , Antibody Formation/genetics , Genetic Code , Inosine/genetics , Inosine/metabolism , Humans
17.
Ann Rheum Dis ; 83(1): 72-87, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-37775153

ABSTRACT

OBJECTIVES: To investigate the effect of the L-arginine metabolism on arthritis and inflammation-mediated bone loss. METHODS: L-arginine was applied to three arthritis models (collagen-induced arthritis, serum-induced arthritis and human TNF transgenic mice). Inflammation was assessed clinically and histologically, while bone changes were quantified by µCT and histomorphometry. In vitro, effects of L-arginine on osteoclast differentiation were analysed by RNA-seq and mass spectrometry (MS). Seahorse, Single Cell ENergetIc metabolism by profilIng Translation inHibition and transmission electron microscopy were used for detecting metabolic changes in osteoclasts. Moreover, arginine-associated metabolites were measured in the serum of rheumatoid arthritis (RA) and pre-RA patients. RESULTS: L-arginine inhibited arthritis and bone loss in all three models and directly blocked TNFα-induced murine and human osteoclastogenesis. RNA-seq and MS analyses indicated that L-arginine switched glycolysis to oxidative phosphorylation in inflammatory osteoclasts leading to increased ATP production, purine metabolism and elevated inosine and hypoxanthine levels. Adenosine deaminase inhibitors blocking inosine and hypoxanthine production abolished the inhibition of L-arginine on osteoclastogenesis in vitro and in vivo. Altered arginine levels were also found in RA and pre-RA patients. CONCLUSION: Our study demonstrated that L-arginine ameliorates arthritis and bone erosion through metabolic reprogramming and perturbation of purine metabolism in osteoclasts.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Bone Resorption , Humans , Mice , Animals , Osteoclasts , Arthritis, Rheumatoid/pathology , Arthritis, Experimental/pathology , Inflammation/metabolism , Mice, Transgenic , Arginine/pharmacology , Inosine/metabolism , Inosine/pharmacology , Hypoxanthines/metabolism , Hypoxanthines/pharmacology , Purines/pharmacology
18.
Structure ; 32(1): 83-96.e4, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38042148

ABSTRACT

Nucleobases such as inosine have been extensively utilized to map direct contacts by proteins in the DNA groove. Their deployment as targeted probes of dynamics and hydration, which are dominant thermodynamic drivers of affinity and specificity, has been limited by a paucity of suitable experimental models. We report a joint crystallographic, thermodynamic, and computational study of the bidentate complex of the arginine side chain with a Watson-Crick guanine (Arg×GC), a highly specific configuration adopted by major transcription factors throughout the eukaryotic branches in the Tree of Life. Using the ETS-family factor PU.1 as a high-resolution structural framework, inosine substitution for guanine resulted in a sharp dissection of conformational dynamics and hydration and elucidated their role in the DNA specificity of PU.1. Our work suggests an under-exploited utility of modified nucleobases in untangling the structural thermodynamics of interactions, such as the Arg×GC motif, where direct and indirect readout are tightly integrated.


Subject(s)
Proto-Oncogene Proteins , Transcription Factors , Transcription Factors/metabolism , Binding Sites , Protein Binding , Proto-Oncogene Proteins/chemistry , Thermodynamics , DNA/metabolism , Guanine , Inosine/metabolism , Nucleic Acid Conformation
19.
Mol Nutr Food Res ; 68(2): e2300115, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38039425

ABSTRACT

BACKGROUND: Oral inosine loading is a new method to evaluate the effects of purine on urate metabolism. However, individuals respond differently to acute purine intake, and the effects on the metabolism of other purines remain to be explored. METHODS: 35 male participants are recruited. Participants received 500 mg of inosine orally after an overnight fast, and blood and urine samples are collected before and at various time points over 180 min after inosine administration. RESULTS: The serum urate concentration is significantly different between the hyperuricemia (n = 14) and non-hyperuricemia (n = 16) groups before inosine intake, but there is no in urate change after inosine intake. When grouped according to the baseline estimated glomerular filtration rate (eGFR), the increase in urate level in the high-eGFR group is significantly higher than that in the low-eGFR group (p  =  0.047). The high-eGFR group showed higher levels of serum xanthine and xanthine oxidase (XOD), the key enzyme in urate synthesis, after inosine loading (p < 0.01). CONCLUSIONS: The increase in urate level is positively related to eGFR after oral acute inosine administration, which may have been due to a higher level of XOD.


Subject(s)
Hyperuricemia , Uric Acid , Humans , Male , Purines/metabolism , Hyperuricemia/drug therapy , Inosine/metabolism , Metabolic Networks and Pathways , China
20.
J Agric Food Chem ; 71(50): 20210-20221, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38079219

ABSTRACT

Inosine is widely used in food, chemical, and medicine. This study developed Bacillus licheniformis into an inosine hyperproducer through systems metabolic engineering. First, purine metabolism was activated by deleting inhibitors PurR and YabJ and overexpressing the pur operon. Then, the 5-phosphoribosyl-1-pyrophosphate (PRPP) supply was increased by optimizing the glucose transport system and pentose phosphate pathway, increasing the inosine titer by 97% and decreasing the titers of byproducts by 36%. Next, to prevent the degradation of inosine, genes deoD and pupG coding purine nucleoside phosphorylase were deleted, accumulating 0.91 g/L inosine in the culture medium. Additionally, the downregulation of adenosine 5'-monophosphate (AMP) synthesis pathway increased the inosine titer by 409%. Importantly, enhancing the glycine and aspartate supply increased the inosine titer by 298%. Finally, the guanosine synthesis pathway was blocked, leading to strain IR-8-2 producing 27.41 g/L inosine with a 0.46 g inosine/g glucose yield and a 0.38 g/(L·h) productivity in a shake flask.


Subject(s)
Bacillus licheniformis , Bacillus licheniformis/genetics , Bacillus licheniformis/metabolism , Metabolic Engineering , Inosine/metabolism , Biosynthetic Pathways , Glucose
SELECTION OF CITATIONS
SEARCH DETAIL
...