Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Cell ; 177(3): 711-721.e8, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30982603

ABSTRACT

Yeast ataxin-2, also known as Pbp1, senses the activity state of mitochondria in order to regulate TORC1. A domain of Pbp1 required to adapt cells to mitochondrial activity is of low sequence complexity. The low-complexity (LC) domain of Pbp1 forms labile, cross-ß polymers that facilitate phase transition of the protein into liquid-like or gel-like states. Phase transition for other LC domains is reliant upon widely distributed aromatic amino acids. In place of tyrosine or phenylalanine residues prototypically used for phase separation, Pbp1 contains 24 similarly disposed methionine residues. Here, we show that the Pbp1 methionine residues are sensitive to hydrogen peroxide (H2O2)-mediated oxidation in vitro and in living cells. Methionine oxidation melts Pbp1 liquid-like droplets in a manner reversed by methionine sulfoxide reductase enzymes. These observations explain how reversible formation of labile polymers by the Pbp1 LC domain enables the protein to function as a sensor of cellular redox state.


Subject(s)
Carrier Proteins/metabolism , Methionine/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Hydrogen Peroxide/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Methionine/metabolism , Methionine Sulfoxide Reductases/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mutagenesis, Site-Directed , Oxidation-Reduction , Oxidative Stress/drug effects , Phase Transition , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
2.
Cell ; 177(3): 697-710.e17, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30982600

ABSTRACT

Yeast ataxin-2, also known as Pbp1 (polyA binding protein-binding protein 1), is an intrinsically disordered protein implicated in stress granule formation, RNA biology, and neurodegenerative disease. To understand the endogenous function of this protein, we identify Pbp1 as a dedicated regulator of TORC1 signaling and autophagy under conditions that require mitochondrial respiration. Pbp1 binds to TORC1 specifically during respiratory growth, but utilizes an additional methionine-rich, low complexity (LC) region to inhibit TORC1. This LC region causes phase separation, forms reversible fibrils, and enables self-association into assemblies required for TORC1 inhibition. Mutants that weaken phase separation in vitro exhibit reduced capacity to inhibit TORC1 and induce autophagy. Loss of Pbp1 leads to mitochondrial dysfunction and reduced fitness during nutritional stress. Thus, Pbp1 forms a condensate in response to respiratory status to regulate TORC1 signaling.


Subject(s)
Carrier Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction , Amino Acid Sequence , Autophagy/drug effects , Carrier Proteins/chemistry , Carrier Proteins/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Methionine/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Domains , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction/drug effects , Sirolimus/pharmacology
3.
Cell ; 169(5): 824-835.e14, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525753

ABSTRACT

Maintenance of proper levels of the methyl donor S-adenosylmethionine (SAM) is critical for a wide variety of biological processes. We demonstrate that the N6-adenosine methyltransferase METTL16 regulates expression of human MAT2A, which encodes the SAM synthetase expressed in most cells. Upon SAM depletion by methionine starvation, cells induce MAT2A expression by enhanced splicing of a retained intron. Induction requires METTL16 and its methylation substrate, a vertebrate conserved hairpin (hp1) in the MAT2A 3' UTR. Increasing METTL16 occupancy on the MAT2A 3' UTR is sufficient to induce efficient splicing. We propose that, under SAM-limiting conditions, METTL16 occupancy on hp1 increases due to inefficient enzymatic turnover, which promotes MAT2A splicing. We further show that METTL16 is the long-unknown methyltransferase for the U6 spliceosomal small nuclear RNA (snRNA). These observations suggest that the conserved U6 snRNA methyltransferase evolved an additional function in vertebrates to regulate SAM homeostasis.


Subject(s)
Introns , Methionine Adenosyltransferase/genetics , Methyltransferases/metabolism , RNA Splicing , S-Adenosylmethionine/metabolism , Animals , Base Sequence , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Inverted Repeat Sequences , Methionine Adenosyltransferase/chemistry , Methylation , Methyltransferases/chemistry , Schizosaccharomyces/metabolism
4.
Mol Cell ; 82(1): 60-74.e5, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34995509

ABSTRACT

Acetyl-CoA is a key intermediate situated at the intersection of many metabolic pathways. The reliance of histone acetylation on acetyl-CoA enables the coordination of gene expression with metabolic state. Abundant acetyl-CoA has been linked to the activation of genes involved in cell growth or tumorigenesis through histone acetylation. However, the role of histone acetylation in transcription under low levels of acetyl-CoA remains poorly understood. Here, we use a yeast starvation model to observe the dramatic alteration in the global occupancy of histone acetylation following carbon starvation; the location of histone acetylation marks shifts from growth-promoting genes to gluconeogenic and fat metabolism genes. This reallocation is mediated by both the histone deacetylase Rpd3p and the acetyltransferase Gcn5p, a component of the SAGA transcriptional coactivator. Our findings reveal an unexpected switch in the specificity of histone acetylation to promote pathways that generate acetyl-CoA for oxidation when acetyl-CoA is limiting.


Subject(s)
Gluconeogenesis , Glucose/deficiency , Histones/metabolism , Lipid Metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae/metabolism , Acetyl Coenzyme A/metabolism , Acetylation , Gene Expression Regulation, Fungal , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Lipid Metabolism/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
5.
Cell ; 159(7): 1591-602, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525877

ABSTRACT

Acetyl-CoA represents a central node of carbon metabolism that plays a key role in bioenergetics, cell proliferation, and the regulation of gene expression. Highly glycolytic or hypoxic tumors must produce sufficient quantities of this metabolite to support cell growth and survival under nutrient-limiting conditions. Here, we show that the nucleocytosolic acetyl-CoA synthetase enzyme, ACSS2, supplies a key source of acetyl-CoA for tumors by capturing acetate as a carbon source. Despite exhibiting no gross deficits in growth or development, adult mice lacking ACSS2 exhibit a significant reduction in tumor burden in two different models of hepatocellular carcinoma. ACSS2 is expressed in a large proportion of human tumors, and its activity is responsible for the majority of cellular acetate uptake into both lipids and histones. These observations may qualify ACSS2 as a targetable metabolic vulnerability of a wide spectrum of tumors.


Subject(s)
Acetate-CoA Ligase/metabolism , Acetates/metabolism , Neoplasms/metabolism , Acetate-CoA Ligase/analysis , Acetate-CoA Ligase/genetics , Acetyl Coenzyme A/metabolism , Animals , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , Mice , Neoplasms/chemistry , Neoplasms/pathology , Positron-Emission Tomography , Triple Negative Breast Neoplasms/chemistry , Triple Negative Breast Neoplasms/pathology
6.
Cell ; 159(7): 1603-14, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25525878

ABSTRACT

Glioblastomas and brain metastases are highly proliferative brain tumors with short survival times. Previously, using (13)C-NMR analysis of brain tumors resected from patients during infusion of (13)C-glucose, we demonstrated that there is robust oxidation of glucose in the citric acid cycle, yet glucose contributes less than 50% of the carbons to the acetyl-CoA pool. Here, we show that primary and metastatic mouse orthotopic brain tumors have the capacity to oxidize [1,2-(13)C]acetate and can do so while simultaneously oxidizing [1,6-(13)C]glucose. The tumors do not oxidize [U-(13)C]glutamine. In vivo oxidation of [1,2-(13)C]acetate was validated in brain tumor patients and was correlated with expression of acetyl-CoA synthetase enzyme 2, ACSS2. Together, the data demonstrate a strikingly common metabolic phenotype in diverse brain tumors that includes the ability to oxidize acetate in the citric acid cycle. This adaptation may be important for meeting the high biosynthetic and bioenergetic demands of malignant growth.


Subject(s)
Acetate-CoA Ligase/metabolism , Acetates/metabolism , Brain Neoplasms/metabolism , Citric Acid Cycle , Glioblastoma/metabolism , Acetate-CoA Ligase/genetics , Animals , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Disease Models, Animal , Glioblastoma/pathology , Glutamic Acid/metabolism , Humans , Mice , Neoplasm Metastasis/pathology
7.
Mol Cell ; 81(18): 3659-3664, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34547228

ABSTRACT

To celebrate our Focus Issue, we asked a selection of researchers working on different aspects of metabolism what they are excited about and what is still to come. They discuss emerging concepts, unanswered questions, things to consider, and technologies that are enabling new discoveries, as well as developing and integrating approaches to drive the field forward.


Subject(s)
Metabolism/physiology , Research/trends , Humans , Research Personnel
8.
Cell ; 154(2): 403-15, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870128

ABSTRACT

Autophagy is a process of cellular self-digestion induced by various forms of starvation. Although nitrogen deficit is a common trigger, some yeast cells induce autophagy upon switch from a rich to minimal media without nitrogen starvation. We show that the amino acid methionine is sufficient to inhibit such non-nitrogen-starvation (NNS)-induced autophagy. Methionine boosts synthesis of the methyl donor, S-adenosylmethionine (SAM). SAM inhibits autophagy and promotes growth through the action of the methyltransferase Ppm1p, which modifies the catalytic subunit of PP2A in tune with SAM levels. Methylated PP2A promotes dephosphorylation of Npr2p, a component of a conserved complex that regulates NNS autophagy and other growth-related processes. Thus, methionine and SAM levels represent a critical gauge of amino acid availability that is sensed via the methylation of PP2A to reciprocally regulate cell growth and autophagy.


Subject(s)
Autophagy , Methionine/metabolism , Protein Phosphatase 2/metabolism , S-Adenosylmethionine/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Protein Methyltransferases/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/metabolism
9.
Cell ; 154(2): 416-29, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870129

ABSTRACT

Protein translation is an energetically demanding process that must be regulated in response to changes in nutrient availability. Herein, we report that intracellular methionine and cysteine availability directly controls the thiolation status of wobble-uridine (U34) nucleotides present on lysine, glutamine, or glutamate tRNAs to regulate cellular translational capacity and metabolic homeostasis. tRNA thiolation is important for growth under nutritionally challenging environments and required for efficient translation of genes enriched in lysine, glutamine, and glutamate codons, which are enriched in proteins important for translation and growth-specific processes. tRNA thiolation is downregulated during sulfur starvation in order to decrease sulfur consumption and growth, and its absence leads to a compensatory increase in enzymes involved in methionine, cysteine, and lysine biosynthesis. Thus, tRNA thiolation enables cells to modulate translational capacity according to the availability of sulfur amino acids, establishing a functional significance for this conserved tRNA nucleotide modification in cell growth control.


Subject(s)
Amino Acids, Sulfur/metabolism , Protein Biosynthesis , RNA, Transfer/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Uridine/metabolism , Down-Regulation , RNA, Transfer/chemistry , Saccharomyces cerevisiae/growth & development
10.
Mol Cell ; 79(5): 758-767.e6, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32755596

ABSTRACT

During proteotoxic stress, bacteria maintain critical processes like DNA replication while removing misfolded proteins, which are degraded by the Lon protease. Here, we show that in Caulobacter crescentus Lon controls deoxyribonucleoside triphosphate (dNTP) pools during stress through degradation of the transcription factor CcrM. Elevated dNTP/nucleotide triphosphate (NTP) ratios in Δlon cells protects them from deletion of otherwise essential deoxythymidine triphosphate (dTTP)-producing pathways and shields them from hydroxyurea-induced loss of dNTPs. Increased dNTP production in Δlon results from higher expression of ribonucleotide reductase driven by increased CcrM. We show that misfolded proteins can stabilize CcrM by competing for limited protease and that Lon-dependent control of dNTPs improves fitness during protein misfolding conditions. We propose that linking dNTP production with availability of Lon allows Caulobacter to maintain replication capacity when misfolded protein burden increases, such as during rapid growth. Because Lon recognizes misfolded proteins regardless of the stress, this mechanism allows for response to a variety of unanticipated conditions.


Subject(s)
Caulobacter crescentus/metabolism , Nucleotides/metabolism , Protease La/metabolism , Protein Folding , Bacterial Proteins/metabolism , Caulobacter crescentus/enzymology , DNA Transposable Elements , Dideoxynucleosides/metabolism , Gene Expression Regulation, Bacterial , Nucleotide Deaminases/genetics , Nucleotide Deaminases/metabolism , Ribonucleotide Reductases/metabolism , Stress, Physiological , Transcription Factors/metabolism , Up-Regulation
11.
Mol Cell ; 78(2): 210-223.e8, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32208170

ABSTRACT

S-adenosylmethionine (SAM) is the methyl-donor substrate for DNA and histone methyltransferases that regulate epigenetic states and subsequent gene expression. This metabolism-epigenome link sensitizes chromatin methylation to altered SAM abundance, yet the mechanisms that allow organisms to adapt and protect epigenetic information during life-experienced fluctuations in SAM availability are unknown. We identified a robust response to SAM depletion that is highlighted by preferential cytoplasmic and nuclear mono-methylation of H3 Lys 9 (H3K9) at the expense of broad losses in histone di- and tri-methylation. Under SAM-depleted conditions, H3K9 mono-methylation preserves heterochromatin stability and supports global epigenetic persistence upon metabolic recovery. This unique chromatin response was robust across the mouse lifespan and correlated with improved metabolic health, supporting a significant role for epigenetic adaptation to SAM depletion in vivo. Together, these studies provide evidence for an adaptive response that enables epigenetic persistence to metabolic stress.


Subject(s)
DNA Methylation/genetics , Heterochromatin/genetics , Metabolome/genetics , S-Adenosylmethionine/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , HCT116 Cells , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Methionine/genetics , Mice , Protein Processing, Post-Translational/genetics , Proteomics/methods
12.
Mol Cell ; 73(6): 1115-1126.e6, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30772176

ABSTRACT

Dysregulation of chromatin methylation is associated with defects in cellular differentiation as well as a variety of cancers. How cells regulate the opposing activities of histone methyltransferase and demethylase enzymes to set the methylation status of the epigenome for proper control of gene expression and metabolism remains poorly understood. Here, we show that loss of methylation of the major phosphatase PP2A in response to methionine starvation activates the demethylation of histones through hyperphosphorylation of specific demethylase enzymes. In parallel, this regulatory mechanism enables cells to preserve SAM by increasing SAH to limit SAM consumption by methyltransferase enzymes. Mutants lacking the PP2A methyltransferase or the effector H3K36 demethylase Rph1 exhibit elevated SAM levels and are dependent on cysteine due to reduced capacity to sink the methyl groups of SAM. Therefore, PP2A directs the methylation status of histones by regulating the phosphorylation status of histone demethylase enzymes in response to SAM levels.


Subject(s)
Chromatin/metabolism , DNA Methylation , Histones/metabolism , Protein Phosphatase 2/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Chromatin/genetics , Dealkylation , Gene Expression Regulation, Fungal , Histone Demethylases/genetics , Histone Demethylases/metabolism , Methylation , Mutation , Protein Binding , Protein Phosphatase 2/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , S-Adenosylmethionine/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
13.
J Cell Sci ; 137(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38842578

ABSTRACT

An important mechanism of gene expression regulation is the epigenetic modification of histones. The cofactors and substrates for these modifications are often intermediary metabolites, and it is becoming increasingly clear that the metabolic and nutritional state of cells can influence these marks. These connections between the balance of metabolites, histone modifications and downstream transcriptional changes comprise a metabolic signaling program that can enable cells to adapt to changes in nutrient availability. Beyond acetylation, there is evidence now that histones can be modified by other acyl groups. In this Cell Science at a Glance article and the accompanying poster, we focus on these histone acylation modifications and provide an overview of the players that govern these acylations and their connections with metabolism.


Subject(s)
Histones , Protein Processing, Post-Translational , Animals , Humans , Acylation , Epigenesis, Genetic , Histones/metabolism
14.
Annu Rev Cell Dev Biol ; 28: 59-87, 2012.
Article in English | MEDLINE | ID: mdl-22578140

ABSTRACT

For unicellular organisms, the decision to enter the cell cycle can be viewed most fundamentally as a metabolic problem. A cell must assess its nutritional and metabolic status to ensure it can synthesize sufficient biomass to produce a new daughter cell. The cell must then direct the appropriate metabolic outputs to ensure completion of the division process. Herein, we discuss the changes in metabolism that accompany entry to, and exit from, the cell cycle for the unicellular eukaryote Saccharomyces cerevisiae. Studies of budding yeast under continuous, slow-growth conditions have provided insights into the essence of these metabolic changes at unprecedented temporal resolution. Some of these mechanisms by which cell growth and proliferation are coordinated with metabolism are likely to be conserved in multicellular organisms. An improved understanding of the metabolic basis of cell cycle control promises to reveal fundamental principles governing tumorigenesis, metazoan development, niche expansion, and many additional aspects of cell and organismal growth control.


Subject(s)
Cell Cycle , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Energy Metabolism , Genes, Fungal , Metabolic Networks and Pathways , Mitosis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology
15.
PLoS Genet ; 19(5): e1010774, 2023 05.
Article in English | MEDLINE | ID: mdl-37216416

ABSTRACT

Pbp1 (poly(A)-binding protein-binding protein 1) is a cytoplasmic stress granule marker that is capable of forming condensates that function in the negative regulation of TORC1 signaling under respiratory conditions. Polyglutamine expansions in its mammalian ortholog ataxin-2 lead to spinocerebellar dysfunction due to toxic protein aggregation. Here, we show that loss of Pbp1 in S. cerevisiae leads to decreased amounts of mRNAs and mitochondrial proteins which are targets of Puf3, a member of the PUF (Pumilio and FBF) family of RNA-binding proteins. We found that Pbp1 supports the translation of Puf3-target mRNAs in respiratory conditions, such as those involved in the assembly of cytochrome c oxidase and subunits of mitochondrial ribosomes. We further show that Pbp1 and Puf3 interact through their respective low complexity domains, which is required for Puf3-target mRNA translation. Our findings reveal a key role for Pbp1-containing assemblies in enabling the translation of mRNAs critical for mitochondrial biogenesis and respiration. They may further explain prior associations of Pbp1/ataxin-2 with RNA, stress granule biology, mitochondrial function, and neuronal health.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Animals , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ataxin-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Organelle Biogenesis , RNA-Binding Proteins/metabolism , Mammals/genetics , Carrier Proteins/genetics
16.
Mol Cell ; 66(2): 180-193.e8, 2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28366644

ABSTRACT

S-adenosylmethionine (SAM) is the methyl donor for biological methylation modifications that regulate protein and nucleic acid functions. Here, we show that methylation of a phospholipid, phosphatidylethanolamine (PE), is a major consumer of SAM. The induction of phospholipid biosynthetic genes is accompanied by induction of the enzyme that hydrolyzes S-adenosylhomocysteine (SAH), a product and inhibitor of methyltransferases. Beyond its function for the synthesis of phosphatidylcholine (PC), the methylation of PE facilitates the turnover of SAM for the synthesis of cysteine and glutathione through transsulfuration. Strikingly, cells that lack PE methylation accumulate SAM, which leads to hypermethylation of histones and the major phosphatase PP2A, dependency on cysteine, and sensitivity to oxidative stress. Without PE methylation, particular sites on histones then become methyl sinks to enable the conversion of SAM to SAH. These findings reveal an unforeseen metabolic function for phospholipid and histone methylation intrinsic to the life of a cell.


Subject(s)
Histones/metabolism , Phosphatidylethanolamines/metabolism , Protein Processing, Post-Translational , S-Adenosylmethionine/metabolism , Saccharomyces cerevisiae/metabolism , Cysteine/metabolism , Energy Metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Lysine/metabolism , Methylation , Mutation , Oxidative Stress , Phosphatidylcholines/metabolism , Phosphatidylethanolamine N-Methyltransferase/genetics , Phosphatidylethanolamine N-Methyltransferase/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , S-Adenosylhomocysteine/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Time Factors , Transcription, Genetic
17.
Proc Natl Acad Sci U S A ; 117(46): 28727-28734, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33144500

ABSTRACT

A methionine-rich low complexity (LC) domain is found within a C-terminal region of the TDP43 RNA-binding protein. Self-association of this domain leads to the formation of labile cross-ß polymers and liquid-like droplets. Treatment with H2O2 caused phenomena of methionine oxidation and droplet melting that were reversed upon exposure of the oxidized protein to methionine sulfoxide reductase enzymes. Morphological features of the cross-ß polymers were revealed by H2O2-mediated footprinting. Equivalent TDP43 LC domain footprints were observed in polymerized hydrogels, liquid-like droplets, and living cells. The ability of H2O2 to impede cross-ß polymerization was abrogated by the prominent M337V amyotrophic lateral sclerosis-causing mutation. These observations may offer insight into the biological role of TDP43 in facilitating synapse-localized translation as well as aberrant aggregation of the protein in neurodegenerative diseases.


Subject(s)
Ataxin-2/metabolism , DNA-Binding Proteins/metabolism , Amino Acid Sequence , Conserved Sequence , HEK293 Cells , Humans , Polymerization , Protein Domains , Reactive Oxygen Species/metabolism
18.
Mol Cell ; 53(3): 363-4, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24507713

ABSTRACT

In this issue of Molecular Cell, Hendriks et al. (2014) uncover extensive oscillations in global gene expression during C. elegans development, in synchrony with the molting cycle.


Subject(s)
Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental , Models, Genetic , Animals
19.
Nucleic Acids Res ; 48(1): 486-499, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31745563

ABSTRACT

Cross-species pathway transplantation enables insight into a biological process not possible through traditional approaches. We replaced the enzymes catalyzing the entire Saccharomyces cerevisiae adenine de novo biosynthesis pathway with the human pathway. While the 'humanized' yeast grew in the absence of adenine, it did so poorly. Dissection of the phenotype revealed that PPAT, the human ortholog of ADE4, showed only partial function whereas all other genes complemented fully. Suppressor analysis revealed other pathways that play a role in adenine de-novo pathway regulation. Phylogenetic analysis pointed to adaptations of enzyme regulation to endogenous metabolite level 'setpoints' in diverse organisms. Using DNA shuffling, we isolated specific amino acids combinations that stabilize the human protein in yeast. Thus, using adenine de novo biosynthesis as a proof of concept, we suggest that the engineering methods used in this study as well as the debugging strategies can be utilized to transplant metabolic pathway from any origin into yeast.


Subject(s)
Adenine/biosynthesis , Biosynthetic Pathways/genetics , Carboxy-Lyases/genetics , Chromosomes, Artificial, Human/chemistry , Peptide Synthases/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , CRISPR-Cas Systems , Carboxy-Lyases/metabolism , Chromosomes, Artificial, Human/metabolism , Genetic Complementation Test , Genetic Engineering/methods , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Peptide Synthases/metabolism , Phylogeny , Plasmids/chemistry , Plasmids/metabolism , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
20.
Proc Natl Acad Sci U S A ; 116(44): 22229-22236, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611405

ABSTRACT

Fumarylacetoacetate hydrolase (FAH) is the last enzyme in tyrosine catabolism, and mutations in the FAH gene are associated with hereditary tyrosinemia type I (HT1 or TYRSN1) in humans. In a behavioral screen of N-ethyl-N-nitrosourea mutagenized mice we identified a mutant line which we named "swingshift" (swst, MGI:3611216) with a nonsynonymous point mutation (N68S) in Fah that caused age-dependent disruption of sleep-wake patterns. Mice homozygous for the mutation had an earlier onset of activity (several hours before lights off) and a reduction in total activity and body weight when compared with wild-type or heterozygous mice. Despite abnormal behavioral entrainment to light-dark cycles, there were no differences in the period or phase of the central clock in mutant mice, indicating a defect downstream of the suprachiasmatic nucleus. Interestingly, these behavioral phenotypes became milder as the mice grew older and were completely rescued by the administration of NTBC [2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione], an inhibitor of 4-hydroxyphenylpyruvate dioxygenase, which is upstream of FAH. Mechanistically, the swst mutation had no effect on the enzymatic activity of FAH, but rather promoted the degradation of the mutant protein. This led to reduced FAH protein levels and enzymatic activity in the liver and kidney (but not the brain or fibroblasts) of homozygous mice. In addition, plasma tyrosine-but not methionine, phenylalanine, or succinylacetone-increased in homozygous mice, suggesting that swst mutants provide a model of mild, chronic HT1.


Subject(s)
Circadian Rhythm , Hydrolases/genetics , Mutation , Sleep , Tyrosinemias/genetics , 4-Hydroxyphenylpyruvate Dioxygenase/antagonists & inhibitors , Animals , Cells, Cultured , Cyclohexanones/therapeutic use , Enzyme Inhibitors/therapeutic use , Enzyme Stability , HEK293 Cells , Homozygote , Humans , Hydrolases/deficiency , Hydrolases/metabolism , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Nitrobenzoates/therapeutic use , Organ Specificity , Suprachiasmatic Nucleus/metabolism , Tyrosinemias/drug therapy , Tyrosinemias/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL