Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell ; 186(9): 1968-1984.e20, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37040760

ABSTRACT

Somatic mutations in nonmalignant tissues accumulate with age and injury, but whether these mutations are adaptive on the cellular or organismal levels is unclear. To interrogate genes in human metabolic disease, we performed lineage tracing in mice harboring somatic mosaicism subjected to nonalcoholic steatohepatitis (NASH). Proof-of-concept studies with mosaic loss of Mboat7, a membrane lipid acyltransferase, showed that increased steatosis accelerated clonal disappearance. Next, we induced pooled mosaicism in 63 known NASH genes, allowing us to trace mutant clones side by side. This in vivo tracing platform, which we coined MOSAICS, selected for mutations that ameliorate lipotoxicity, including mutant genes identified in human NASH. To prioritize new genes, additional screening of 472 candidates identified 23 somatic perturbations that promoted clonal expansion. In validation studies, liver-wide deletion of Tbx3, Bcl6, or Smyd2 resulted in protection against hepatic steatosis. Selection for clonal fitness in mouse and human livers identifies pathways that regulate metabolic disease.


Subject(s)
Metabolic Diseases , Non-alcoholic Fatty Liver Disease , Animals , Humans , Male , Mice , Histone-Lysine N-Methyltransferase/genetics , Liver/metabolism , Mosaicism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism
2.
EMBO J ; 41(6): e108544, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34850409

ABSTRACT

Since numerous RNAs and RBPs prevalently localize to active chromatin regions, many RNA-binding proteins (RBPs) may be potential transcriptional regulators. RBPs are generally thought to regulate transcription via noncoding RNAs. Here, we describe a distinct, dual mechanism of transcriptional regulation by the previously uncharacterized tRNA-modifying enzyme, hTrmt13. On one hand, hTrmt13 acts in the cytoplasm to catalyze 2'-O-methylation of tRNAs, thus regulating translation in a manner depending on its tRNA-modification activity. On the other hand, nucleus-localized hTrmt13 directly binds DNA as a transcriptional co-activator of key epithelial-mesenchymal transition factors, thereby promoting cell migration independent of tRNA-modification activity. These dual functions of hTrmt13 are mutually exclusive, as it can bind either DNA or tRNA through its CHHC zinc finger domain. Finally, we find that hTrmt13 expression is tightly associated with poor prognosis and survival in diverse cancer patients. Our discovery of the noncatalytic roles of an RNA-modifying enzyme provides a new perspective for understanding epitranscriptomic regulation.


Subject(s)
RNA, Transfer , tRNA Methyltransferases , Humans , Methylation , RNA/metabolism , RNA Processing, Post-Transcriptional , RNA, Transfer/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
3.
J Biol Chem ; 299(5): 104704, 2023 05.
Article in English | MEDLINE | ID: mdl-37059185

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are essential components for mRNA translation. Two sets of aaRSs are required for cytoplasmic and mitochondrial translation in vertebrates. Interestingly, TARSL2 is a recently evolved duplicated gene of TARS1 (encoding cytoplasmic threonyl-tRNA synthetase) and represents the only duplicated aaRS gene in vertebrates. Although TARSL2 retains the canonical aminoacylation and editing activities in vitro, whether it is a true tRNA synthetase for mRNA translation in vivo is unclear. In this study, we showed that Tars1 is an essential gene since homozygous Tars1 KO mice were lethal. In contrast, when Tarsl2 was deleted in mice and zebrafish, neither the abundance nor the charging levels of tRNAThrs were changed, indicating that cells relied on Tars1 but not on Tarsl2 for mRNA translation. Furthermore, Tarsl2 deletion did not influence the integrity of the multiple tRNA synthetase complex, suggesting that Tarsl2 is a peripheral member of the multiple tRNA synthetase complex. Finally, we observed that Tarsl2-deleted mice exhibited severe developmental retardation, elevated metabolic capacity, and abnormal bone and muscle development after 3 weeks. Collectively, these data suggest that, despite its intrinsic activity, loss of Tarsl2 has little influence on protein synthesis but does affect mouse development.


Subject(s)
Amino Acyl-tRNA Synthetases , Protein Biosynthesis , Threonine-tRNA Ligase , Animals , Mice , Amino Acyl-tRNA Synthetases/metabolism , RNA, Transfer/metabolism , Threonine-tRNA Ligase/genetics , Threonine-tRNA Ligase/metabolism , Zebrafish/genetics , Zebrafish/metabolism
4.
Hepatology ; 78(4): 1133-1148, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37039560

ABSTRACT

BACKGROUND AND AIMS: The liver is remarkably regenerative and can completely recover even when 80% of its mass is surgically removed. Identification of secreted factors that regulate liver growth would help us understand how organ size and regeneration are controlled but also provide candidate targets to promote regeneration or impair cancer growth. APPROACH AND RESULTS: To enrich for secreted factors that regulate growth control, we induced massive liver overgrowth with either YAP or MYC . Differentially expressed secreted factors were identified in these livers using transcriptomic analysis. To rank candidates by functionality, we performed in vivo CRISPR screening using the Fah knockout model of tyrosinemia. We identified secreted phosphoprotein-2 (SPP2) as a secreted factor that negatively regulates regeneration. Spp2 -deficient mice showed increased survival after acetaminophen poisoning and reduced fibrosis after repeated carbon tetrachloride injections. We examined the impact of SPP2 on bone morphogenetic protein signaling in liver cells and found that SPP2 antagonized bone morphogenetic protein signaling in vitro and in vivo. We also identified cell-surface receptors that interact with SPP2 using a proximity biotinylation assay coupled with mass spectrometry. We showed that SPP2's interactions with integrin family members are in part responsible for some of the regeneration phenotypes. CONCLUSIONS: Using an in vivo CRISPR screening system, we identified SPP2 as a secreted factor that negatively regulates liver regeneration. This study provides ways to identify, validate, and characterize secreted factors in vivo.


Subject(s)
Liver Regeneration , Neoplasms , Mice , Animals , Liver/metabolism , Hepatocytes/metabolism , Signal Transduction
5.
J Chem Phys ; 159(20)2023 Nov 28.
Article in English | MEDLINE | ID: mdl-37991160

ABSTRACT

In the process of high temperature service, the mechanical properties of cutting tools decrease sharply due to the peeling of the protective coating. However, the mechanism of such coating failure remains obscure due to the complicated interaction between atomic structure, temperature, and stress. This dynamic evolution nature demands both large system sizes and accurate description on the atomic scale, raising challenges for existing atomic scale calculation methods. Here, we developed a deep neural network (DNN) potential for Ti-N binary systems based on first-principles study datasets to achieve quantum-accurate large-scale atomic simulation. Compared with empirical interatomic potential based on the embedded-atom-method, the developed DNN-potential can accurately predict lattice constants, phonon properties, and mechanical properties under various thermodynamic conditions. Moreover, for the first time, we present the atomic evolution of the fracture behavior of large-scale rocksalt-structure (B1) TiN systems coupled with temperature and stress conditions. Our study validates that interatomic brittle fractures occur when TiN stretches beyond its tensile yield point. Such simulation of coating fracture and cutting behavior based on large-scale atoms can shed new light on understanding the microstructure and mechanical properties of coating tools under extreme operating conditions.

6.
J Chem Phys ; 159(5)2023 Aug 07.
Article in English | MEDLINE | ID: mdl-37526163

ABSTRACT

DeePMD-kit is a powerful open-source software package that facilitates molecular dynamics simulations using machine learning potentials known as Deep Potential (DP) models. This package, which was released in 2017, has been widely used in the fields of physics, chemistry, biology, and material science for studying atomistic systems. The current version of DeePMD-kit offers numerous advanced features, such as DeepPot-SE, attention-based and hybrid descriptors, the ability to fit tensile properties, type embedding, model deviation, DP-range correction, DP long range, graphics processing unit support for customized operators, model compression, non-von Neumann molecular dynamics, and improved usability, including documentation, compiled binary packages, graphical user interfaces, and application programming interfaces. This article presents an overview of the current major version of the DeePMD-kit package, highlighting its features and technical details. Additionally, this article presents a comprehensive procedure for conducting molecular dynamics as a representative application, benchmarks the accuracy and efficiency of different models, and discusses ongoing developments.

7.
Nucleic Acids Res ; 48(6): 3181-3194, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32047918

ABSTRACT

N 6-Threonylcarbamoyladenosine (t6A) is a universal tRNA modification essential for translational accuracy and fidelity. In human mitochondria, YrdC synthesises an l-threonylcarbamoyl adenylate (TC-AMP) intermediate, and OSGEPL1 transfers the TC-moiety to five tRNAs, including human mitochondrial tRNAThr (hmtRNAThr). Mutation of hmtRNAs, YrdC and OSGEPL1, affecting efficient t6A modification, has been implicated in various human diseases. However, little is known about the tRNA recognition mechanism in t6A formation in human mitochondria. Herein, we showed that OSGEPL1 is a monomer and is unique in utilising C34 as an anti-determinant by studying the contributions of individual bases in the anticodon loop of hmtRNAThr to t6A modification. OSGEPL1 activity was greatly enhanced by introducing G38A in hmtRNAIle or the A28:U42 base pair in a chimeric tRNA containing the anticodon stem of hmtRNASer(AGY), suggesting that sequences of specific hmtRNAs are fine-tuned for different modification levels. Moreover, using purified OSGEPL1, we identified multiple acetylation sites, and OSGEPL1 activity was readily affected by acetylation via multiple mechanisms in vitro and in vivo. Collectively, we systematically elucidated the nucleotide requirement in the anticodon loop of hmtRNAs, and revealed mechanisms involving tRNA sequence optimisation and post-translational protein modification that determine t6A modification levels.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Mitochondria/genetics , Protein Biosynthesis , RNA, Transfer, Thr/genetics , Acetylation , Adenosine/analogs & derivatives , Adenosine/genetics , Anticodon/genetics , Escherichia coli/genetics , GTP-Binding Proteins/genetics , Humans , Mutation , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional/genetics , RNA, Transfer/genetics , RNA-Binding Proteins/genetics
8.
Nucleic Acids Res ; 47(16): 8662-8674, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31287872

ABSTRACT

A typical feature of eukaryotic aminoacyl-tRNA synthetases (aaRSs) is the evolutionary gain of domains at either the N- or C-terminus, which frequently mediating protein-protein interaction. TARSL2 (mouse Tarsl2), encoding a threonyl-tRNA synthetase-like protein (ThrRS-L), is a recently identified aaRS-duplicated gene in higher eukaryotes, with canonical functions in vitro, which exhibits a different N-terminal extension (N-extension) from TARS (encoding ThrRS). We found the first half of the N-extension of human ThrRS-L (hThrRS-L) is homologous to that of human arginyl-tRNA synthetase. Using the N-extension as a probe in a yeast two-hybrid screening, AIMP1/p43 was identified as an interactor with hThrRS-L. We showed that ThrRS-L is a novel component of the mammalian multiple tRNA synthetase complex (MSC), and is reliant on two leucine zippers in the N-extension for MSC-incorporation in humans, and mouse cell lines and muscle tissue. The N-extension was sufficient to target a foreign protein into the MSC. The results from a Tarsl2-deleted cell line showed that it does not mediate MSC integrity. The effect of phosphorylation at various sites of hThrRS-L on its MSC-targeting is also explored. In summary, we revealed that ThrRS-L is a bona fide component of the MSC, which is mediated by a newly evolved N-extension domain.


Subject(s)
Arginine-tRNA Ligase/genetics , Cytokines/genetics , Multienzyme Complexes/genetics , Neoplasm Proteins/genetics , RNA-Binding Proteins/genetics , Threonine-tRNA Ligase/genetics , Amino Acid Sequence , Animals , Arginine-tRNA Ligase/metabolism , Cloning, Molecular , Cytokines/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Humans , Leucine Zippers , Mice , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Neoplasm Proteins/metabolism , Phosphorylation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , RNA-Binding Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Threonine-tRNA Ligase/metabolism , Two-Hybrid System Techniques
9.
Nucleic Acids Res ; 47(6): 3072-3085, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30952159

ABSTRACT

Alanyl-tRNA synthetases (AlaRSs) from three domains of life predominantly rely on a single wobble base pair, G3-U70, of tRNAAla as a major determinant. However, this base pair is divergent in human mitochondrial tRNAAla, but instead with a translocated G5-U68. How human mitochondrial AlaRS (hmtAlaRS) recognizes tRNAAla, in particular, in the acceptor stem region, remains unknown. In the present study, we found that hmtAlaRS is a monomer and recognizes mitochondrial tRNAAla in a G3-U70-independent manner, requiring several elements in the acceptor stem. In addition, we found that hmtAlaRS misactivates noncognate Gly and catalyzes strong transfer RNA (tRNA)-independent pre-transfer editing for Gly. A completely conserved residue outside of the editing active site, Arg663, likely functions as a tRNA translocation determinant to facilitate tRNA entry into the editing domain during editing. Finally, we investigated the effects of the severe infantile-onset cardiomyopathy-associated R592W mutation of hmtAlaRS on the canonical enzymatic activities of hmtAlaRS. Overall, our results provide fundamental information about tRNA recognition and deepen our understanding of translational quality control mechanisms by hmtAlaRS.


Subject(s)
Nucleic Acid Conformation , RNA, Mitochondrial/genetics , RNA, Transfer, Ala/genetics , RNA, Transfer/genetics , Alanine-tRNA Ligase/genetics , Base Pairing/genetics , Catalytic Domain , Escherichia coli/genetics , Humans , Kinetics , Models, Molecular , Substrate Specificity
10.
Nucleic Acids Res ; 46(9): 4662-4676, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29648639

ABSTRACT

Six pathogenic mutations have been reported in human mitochondrial tRNAThr (hmtRNAThr); however, the pathogenic molecular mechanism remains unclear. Previously, we established an activity assay system for human mitochondrial threonyl-tRNA synthetase (hmThrRS). In the present study, we surveyed the structural and enzymatic effects of pathogenic mutations in hmtRNAThr and then focused on m.15915 G > A (G30A) and m.15923A > G (A38G). The harmful evolutionary gain of non-Watson-Crick base pair A29/C41 caused hmtRNAThr to be highly susceptible to mutations disrupting the G30-C40 base pair in various ways; for example, structural integrity maintenance, modification and aminoacylation of tRNAThr, and editing mischarged tRNAThr. A similar phenomenon was observed for hmtRNATrp with an A29/C41 non-Watson-Crick base pair, but not in bovine mtRNAThr with a natural G29-C41 base pair. The A38G mutation caused a severe reduction in Thr-acceptance and editing of hmThrRS. Importantly, A38 is a nucleotide determinant for the t6A modification at A37, which is essential for the coding properties of hmtRNAThr. In summary, our results revealed the crucial role of the G30-C40 base pair in maintaining the proper structure and function of hmtRNAThr because of A29/C41 non-Watson-Crick base pair and explained the molecular outcome of pathogenic G30A and A38G mutations.


Subject(s)
Mutation , RNA, Mitochondrial/chemistry , RNA, Transfer, Thr/chemistry , Anticodon , Base Pairing , Humans , Mitochondria/enzymology , RNA Editing , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Transfer, Thr/genetics , RNA, Transfer, Thr/metabolism , Threonine-tRNA Ligase/metabolism , Transfer RNA Aminoacylation
11.
Nanomaterials (Basel) ; 13(9)2023 May 08.
Article in English | MEDLINE | ID: mdl-37177121

ABSTRACT

The two-dimensional post-transition-metal chalcogenides, particularly indium selenide (InSe), exhibit salient carrier transport properties and evince extensive interest for broad applications. A comprehensive understanding of thermal transport is indispensable for thermal management. However, theoretical predictions on thermal transport in the InSe system are found in disagreement with experimental measurements. In this work, we utilize both the Green-Kubo approach with deep potential (GK-DP), together with the phonon Boltzmann transport equation with density functional theory (BTE-DFT) to investigate the thermal conductivity (κ) of InSe monolayer. The κ calculated by GK-DP is 9.52 W/mK at 300 K, which is in good agreement with the experimental value, while the κ predicted by BTE-DFT is 13.08 W/mK. After analyzing the scattering phase space and cumulative κ by mode-decomposed method, we found that, due to the large energy gap between lower and upper optical branches, the exclusion of four-phonon scattering in BTE-DFT underestimates the scattering phase space of lower optical branches due to large group velocities, and thus would overestimate their contribution to κ. The temperature dependence of κ calculated by GK-DP also demonstrates the effect of higher-order phonon scattering, especially at high temperatures. Our results emphasize the significant role of four-phonon scattering in InSe monolayer, suggesting that combining molecular dynamics with machine learning potential is an accurate and efficient approach to predict thermal transport.

12.
Cell Stem Cell ; 30(5): 665-676.e4, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37146585

ABSTRACT

Although midlobular hepatocytes in zone 2 are a recently identified cellular source for liver homeostasis and regeneration, these cells have not been exclusively fate mapped. We generated an Igfbp2-CreER knockin strain that specifically labels midlobular hepatocytes. During homeostasis over 1 year, zone 2 hepatocytes increased in abundance from occupying 21%-41% of the lobular area. After either pericentral injury with carbon tetrachloride or periportal injury with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), IGFBP2+ cells replenished lost hepatocytes in zones 3 and 1, respectively. IGFBP2+ cells also preferentially contributed to regeneration after 70% partial hepatectomy, as well as liver growth during pregnancy. Because IGFBP2 labeling increased substantially with fasting, we used single nuclear transcriptomics to explore zonation as a function of nutrition, revealing that the zonal division of labor shifts dramatically with fasting. These studies demonstrate the contribution of IGFBP2-labeled zone 2 hepatocytes to liver homeostasis and regeneration.


Subject(s)
Insulin-Like Growth Factor Binding Protein 2 , Liver Regeneration , Liver , Hepatectomy , Hepatocytes , Homeostasis , Insulin-Like Growth Factor Binding Protein 2/metabolism
13.
bioRxiv ; 2023 Mar 21.
Article in English | MEDLINE | ID: mdl-36993727

ABSTRACT

Somatic mutations in non-malignant tissues accumulate with age and insult, but whether these mutations are adaptive on the cellular or organismal levels is unclear. To interrogate mutations found in human metabolic disease, we performed lineage tracing in mice harboring somatic mosaicism subjected to non-alcoholic steatohepatitis (NASH). Proof-of-concept studies with mosaic loss of Mboat7 , a membrane lipid acyltransferase, showed that increased steatosis accelerated clonal disappearance. Next, we induced pooled mosaicism in 63 known NASH genes, allowing us to trace mutant clones side-by-side. This in vivo tracing platform, which we coined MOSAICS, selected for mutations that ameliorate lipotoxicity, including mutant genes identified in human NASH. To prioritize new genes, additional screening of 472 candidates identified 23 somatic perturbations that promoted clonal expansion. In validation studies, liver-wide deletion of Bcl6, Tbx3, or Smyd2 resulted in protection against NASH. Selection for clonal fitness in mouse and human livers identifies pathways that regulate metabolic disease. Highlights: Mosaic Mboat7 mutations that increase lipotoxicity lead to clonal disappearance in NASH. In vivo screening can identify genes that alter hepatocyte fitness in NASH. Mosaic Gpam mutations are positively selected due to reduced lipogenesis. In vivo screening of transcription factors and epifactors identified new therapeutic targets in NASH.

14.
Cell Discov ; 7(1): 98, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34697290

ABSTRACT

The amino acid response (AAR) and unfolded protein response (UPR) pathways converge on eIF2α phosphorylation, which is catalyzed by Gcn2 and Perk, respectively, under different stresses. This close interconnection makes it difficult to specify different functions of AAR and UPR. Here, we generated a zebrafish model in which loss of threonyl-tRNA synthetase (Tars) induces angiogenesis dependent on Tars aminoacylation activity. Comparative transcriptome analysis of the tars-mutant and wild-type embryos with/without Gcn2- or Perk-inhibition reveals that only Gcn2-mediated AAR is activated in the tars-mutants, whereas Perk functions predominantly in normal development. Mechanistic analysis shows that, while a considerable amount of eIF2α is normally phosphorylated by Perk, the loss of Tars causes an accumulation of uncharged tRNAThr, which in turn activates Gcn2, leading to phosphorylation of an extra amount of eIF2α. The partial switchover of kinases for eIF2α largely overwhelms the functions of Perk in normal development. Interestingly, although inhibition of Gcn2 and Perk in this stress condition both can reduce the eIF2α phosphorylation levels, their functional consequences in the regulation of target genes and in the rescue of the angiogenic phenotypes are dramatically different. Indeed, genetic and pharmacological manipulations of these pathways validate that the Gcn2-mediated AAR, but not the Perk-mediated UPR, is required for tars-deficiency induced angiogenesis. Thus, the interconnected AAR and UPR pathways differentially regulate angiogenesis through selective functions and mutual competitions, reflecting the specificity and efficiency of multiple stress response pathways that evolve integrally to enable an organism to sense/respond precisely to various types of stresses.

15.
Sci China Life Sci ; 63(8): 1227-1239, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32189241

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are ubiquitously expressed, essential enzymes, synthesizing aminoacyl-tRNAs for protein synthesis. Functional defects of aaRSs frequently cause various human disorders. Human KARS encodes both cytosolic and mitochondrial lysyl-tRNA synthetases (LysRSs). Previously, two mutations (c.1129G>A and c.517T>C) were identified that led to hearing impairment; however, the underlying biochemical mechanism is unclear. In the present study, we found that the two mutations have no impact on the incorporation of LysRS into the multiple-synthetase complex in the cytosol, but affect the cytosolic LysRS level, its tertiary structure, and cytosolic tRNA aminoacylation in vitro. As for mitochondrial translation, the two mutations have little effect on the steady-state level, mitochondrial targeting, and tRNA binding affinity of mitochondrial LysRS. However, they exhibit striking differences in charging mitochondrial tRNALys, with the c.517T>C mutant being completely deficient in vitro and in vivo. We constructed two yeast genetic models, which are powerful tools to test the in vivo aminoacylation activity of KARS mutations at both the cytosolic and mitochondrial levels. Overall, our data provided biochemical insights into the potentially molecular pathological mechanism of KARS c.1129G>A and c.517T>C mutations and provided yeast genetic bases to investigate other KARS mutations in the future.


Subject(s)
Aminoacylation/genetics , Cytoplasm/genetics , Hearing Loss/genetics , Mitochondria/genetics , RNA, Transfer, Lys/metabolism , Amino Acyl-tRNA Synthetases/genetics , Base Sequence , Catalytic Domain , Gene Expression Regulation , Hearing Loss/metabolism , Humans , Models, Molecular , Mutation , Protein Biosynthesis , Protein Conformation , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL