Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25.725
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 186(1): 63-79.e21, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36608659

RESUMO

Metabolism is deeply intertwined with aging. Effects of metabolic interventions on aging have been explained with intracellular metabolism, growth control, and signaling. Studying chronological aging in yeast, we reveal a so far overlooked metabolic property that influences aging via the exchange of metabolites. We observed that metabolites exported by young cells are re-imported by chronologically aging cells, resulting in cross-generational metabolic interactions. Then, we used self-establishing metabolically cooperating communities (SeMeCo) as a tool to increase metabolite exchange and observed significant lifespan extensions. The longevity of the SeMeCo was attributable to metabolic reconfigurations in methionine consumer cells. These obtained a more glycolytic metabolism and increased the export of protective metabolites that in turn extended the lifespan of cells that supplied them with methionine. Our results establish metabolite exchange interactions as a determinant of cellular aging and show that metabolically cooperating cells can shape the metabolic environment to extend their lifespan.


Assuntos
Longevidade , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Metionina/metabolismo , Transdução de Sinais
2.
Cell ; 177(3): 711-721.e8, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30982603

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Metionina/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/química , Proteínas de Transporte/genética , Peróxido de Hidrogênio/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Metionina/metabolismo , Metionina Sulfóxido Redutases/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mutagênese Sítio-Dirigida , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Transição de Fase , Domínios Proteicos , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
3.
Cell ; 177(3): 697-710.e17, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30982600

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Autofagia/efeitos dos fármacos , Proteínas de Transporte/química , Proteínas de Transporte/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Metionina/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica , Domínios Proteicos , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia
4.
Cell ; 166(5): 1215-1230.e20, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27523608

RESUMO

Methionine-1 (M1)-linked ubiquitin chains regulate the activity of NF-κB, immune homeostasis, and responses to infection. The importance of negative regulators of M1-linked chains in vivo remains poorly understood. Here, we show that the M1-specific deubiquitinase OTULIN is essential for preventing TNF-associated systemic inflammation in humans and mice. A homozygous hypomorphic mutation in human OTULIN causes a potentially fatal autoinflammatory condition termed OTULIN-related autoinflammatory syndrome (ORAS). Four independent OTULIN mouse models reveal that OTULIN deficiency in immune cells results in cell-type-specific effects, ranging from over-production of inflammatory cytokines and autoimmunity due to accumulation of M1-linked polyubiquitin and spontaneous NF-κB activation in myeloid cells to downregulation of M1-polyubiquitin signaling by degradation of LUBAC in B and T cells. Remarkably, treatment with anti-TNF neutralizing antibodies ameliorates inflammation in ORAS patients and rescues mouse phenotypes. Hence, OTULIN is critical for restraining life-threatening spontaneous inflammation and maintaining immune homeostasis.


Assuntos
Doenças Autoimunes/genética , Autoimunidade/genética , Enzimas Desubiquitinantes/metabolismo , Endopeptidases/metabolismo , Inflamação/genética , Animais , Anticorpos Neutralizantes/uso terapêutico , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Linfócitos B/imunologia , Citocinas/metabolismo , Enzimas Desubiquitinantes/genética , Modelos Animais de Doenças , Endopeptidases/genética , Mutação em Linhagem Germinativa , Humanos , Inflamação/imunologia , Inflamação/terapia , Infliximab/uso terapêutico , Metionina/metabolismo , Camundongos , Camundongos Mutantes , Células Mieloides/imunologia , Poliubiquitina/metabolismo , Deleção de Sequência , Síndrome , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
5.
Cell ; 160(1-2): 132-44, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25542313

RESUMO

Dietary restriction (DR) without malnutrition encompasses numerous regimens with overlapping benefits including longevity and stress resistance, but unifying nutritional and molecular mechanisms remain elusive. In a mouse model of DR-mediated stress resistance, we found that sulfur amino acid (SAA) restriction increased expression of the transsulfuration pathway (TSP) enzyme cystathionine γ-lyase (CGL), resulting in increased hydrogen sulfide (H2S) production and protection from hepatic ischemia reperfusion injury. SAA supplementation, mTORC1 activation, or chemical/genetic CGL inhibition reduced H2S production and blocked DR-mediated stress resistance. In vitro, the mitochondrial protein SQR was required for H2S-mediated protection during nutrient/oxygen deprivation. Finally, TSP-dependent H2S production was observed in yeast, worm, fruit fly, and rodent models of DR-mediated longevity. Together, these data are consistent with evolutionary conservation of TSP-mediated H2S as a mediator of DR benefits with broad implications for clinical translation. PAPERFLICK:


Assuntos
Dieta , Sulfeto de Hidrogênio/metabolismo , Animais , Evolução Biológica , Caenorhabditis elegans/fisiologia , Restrição Calórica , Cistationina gama-Liase/metabolismo , Cisteína/metabolismo , Drosophila melanogaster/fisiologia , Feminino , Rim/irrigação sanguínea , Rim/lesões , Expectativa de Vida , Fígado/irrigação sanguínea , Fígado/lesões , Masculino , Metionina/metabolismo , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão , Transdução de Sinais , Estresse Fisiológico , Transcriptoma , Leveduras/fisiologia
6.
Mol Cell ; 82(16): 3045-3060.e11, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35752173

RESUMO

Cancer mortality is primarily a consequence of its metastatic spread. Here, we report that methionine sulfoxide reductase A (MSRA), which can reduce oxidized methionine residues, acts as a suppressor of pancreatic ductal adenocarcinoma (PDA) metastasis. MSRA expression is decreased in the metastatic tumors of PDA patients, whereas MSRA loss in primary PDA cells promotes migration and invasion. Chemoproteomic profiling of pancreatic organoids revealed that MSRA loss results in the selective oxidation of a methionine residue (M239) in pyruvate kinase M2 (PKM2). Moreover, M239 oxidation sustains PKM2 in an active tetrameric state to promote respiration, migration, and metastasis, whereas pharmacological activation of PKM2 increases cell migration and metastasis in vivo. These results demonstrate that methionine residues can act as reversible redox switches governing distinct signaling outcomes and that the MSRA-PKM2 axis serves as a regulatory nexus between redox biology and cancer metabolism to control tumor metastasis.


Assuntos
Carcinoma Ductal Pancreático , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias Pancreáticas , Hormônios Tireóideos/metabolismo , Carcinoma Ductal Pancreático/genética , Humanos , Metionina , Metionina Sulfóxido Redutases/química , Metionina Sulfóxido Redutases/metabolismo , Oxirredução , Neoplasias Pancreáticas/genética , Piruvato Quinase/metabolismo , Proteínas de Ligação a Hormônio da Tireoide , Neoplasias Pancreáticas
7.
Annu Rev Biochem ; 83: 779-812, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24499181

RESUMO

In eukaryotes, the translation initiation codon is generally identified by the scanning mechanism, wherein every triplet in the messenger RNA leader is inspected for complementarity to the anticodon of methionyl initiator transfer RNA (Met-tRNAi). Binding of Met-tRNAi to the small (40S) ribosomal subunit, in a ternary complex (TC) with eIF2-GTP, is stimulated by eukaryotic initiation factor 1 (eIF1), eIF1A, eIF3, and eIF5, and the resulting preinitiation complex (PIC) joins the 5' end of mRNA preactivated by eIF4F and poly(A)-binding protein. RNA helicases remove secondary structures that impede ribosome attachment and subsequent scanning. Hydrolysis of eIF2-bound GTP is stimulated by eIF5 in the scanning PIC, but completion of the reaction is impeded at non-AUG triplets. Although eIF1 and eIF1A promote scanning, eIF1 and possibly the C-terminal tail of eIF1A must be displaced from the P decoding site to permit base-pairing between Met-tRNAi and the AUG codon, as well as to allow subsequent phosphate release from eIF2-GDP. A second GTPase, eIF5B, catalyzes the joining of the 60S subunit to produce an 80S initiation complex that is competent for elongation.


Assuntos
Fator de Iniciação 1 em Eucariotos/metabolismo , Fator de Iniciação 3 em Eucariotos/metabolismo , Fator de Iniciação 5 em Eucariotos/metabolismo , RNA de Transferência de Metionina/genética , Subunidades Ribossômicas Menores de Eucariotos/química , Animais , Pareamento de Bases , Sítios de Ligação , Códon de Iniciação , Guanosina Trifosfato/química , Humanos , Hidrólise , Metionina/química , Ligação Proteica , RNA Helicases/química , Ribossomos/química , Tetrahymena
8.
Cell ; 157(3): 624-35, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24766808

RESUMO

Quantitative views of cellular functions require precise measures of rates of biomolecule production, especially proteins-the direct effectors of biological processes. Here, we present a genome-wide approach, based on ribosome profiling, for measuring absolute protein synthesis rates. The resultant E. coli data set transforms our understanding of the extent to which protein synthesis is precisely controlled to optimize function and efficiency. Members of multiprotein complexes are made in precise proportion to their stoichiometry, whereas components of functional modules are produced differentially according to their hierarchical role. Estimates of absolute protein abundance also reveal principles for optimizing design. These include how the level of different types of transcription factors is optimized for rapid response and how a metabolic pathway (methionine biosynthesis) balances production cost with activity requirements. Our studies reveal how general principles, important both for understanding natural systems and for synthesizing new ones, emerge from quantitative analyses of protein synthesis.


Assuntos
Escherichia coli/metabolismo , Biossíntese de Proteínas , Proteínas de Bactérias/metabolismo , Estudo de Associação Genômica Ampla , Metionina/biossíntese , Complexos Multiproteicos/metabolismo , Ribossomos/metabolismo , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo
9.
Cell ; 156(1-2): 158-69, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24361105

RESUMO

The Arg/N-end rule pathway targets for degradation proteins that bear specific unacetylated N-terminal residues while the Ac/N-end rule pathway targets proteins through their N(α)-terminally acetylated (Nt-acetylated) residues. Here, we show that Ubr1, the ubiquitin ligase of the Arg/N-end rule pathway, recognizes unacetylated N-terminal methionine if it is followed by a hydrophobic residue. This capability of Ubr1 expands the range of substrates that can be targeted for degradation by the Arg/N-end rule pathway because virtually all nascent cellular proteins bear N-terminal methionine. We identified Msn4, Sry1, Arl3, and Pre5 as examples of normal or misfolded proteins that can be destroyed through the recognition of their unacetylated N-terminal methionine. Inasmuch as proteins bearing the Nt-acetylated N-terminal methionine residue are substrates of the Ac/N-end rule pathway, the resulting complementarity of the Arg/N-end rule and Ac/N-end rule pathways enables the elimination of protein substrates regardless of acetylation state of N-terminal methionine in these substrates.


Assuntos
Metionina/metabolismo , Sinais Direcionadores de Proteínas , Proteólise , Sequência de Aminoácidos , Animais , Redes e Vias Metabólicas , Camundongos , Dados de Sequência Molecular , Dobramento de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
10.
Cell ; 156(4): 759-70, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24529378

RESUMO

Diet greatly influences gene expression and physiology. In mammals, elucidating the effects and mechanisms of individual nutrients is challenging due to the complexity of both the animal and its diet. Here, we used an interspecies systems biology approach with Caenorhabditis elegans and two of its bacterial diets, Escherichia coli and Comamonas aquatica, to identify metabolites that affect the animal's gene expression and physiology. We identify vitamin B12 as the major dilutable metabolite provided by Comamonas aq. that regulates gene expression, accelerates development, and reduces fertility but does not affect lifespan. We find that vitamin B12 has a dual role in the animal: it affects development and fertility via the methionine/S-Adenosylmethionine (SAM) cycle and breaks down the short-chain fatty acid propionic acid, preventing its toxic buildup. Our interspecies systems biology approach provides a paradigm for understanding complex interactions between diet and physiology.


Assuntos
Betaproteobacteria/metabolismo , Caenorhabditis elegans/fisiologia , Escherichia coli/metabolismo , Regulação da Expressão Gênica , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Dieta , Redes e Vias Metabólicas , Metionina/metabolismo , Dados de Sequência Molecular , Propionatos/metabolismo , S-Adenosilmetionina/metabolismo , Transcriptoma , Vitamina B 12/metabolismo
11.
Mol Cell ; 81(23): 4876-4890.e7, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34739871

RESUMO

Histone H3.3 lysine-to-methionine substitutions K27M and K36M impair the deposition of opposing chromatin marks, H3K27me3/me2 and H3K36me3/me2. We show that these mutations induce hypotrophic and disorganized eyes in Drosophila eye primordia. Restriction of H3K27me3 spread in H3.3K27M and its redistribution in H3.3K36M result in transcriptional deregulation of PRC2-targeted eye development and of piRNA biogenesis genes, including krimp. Notably, both mutants promote redistribution of H3K36me2 away from repetitive regions into active genes, which associate with retrotransposon de-repression in eye discs. Aberrant expression of krimp represses LINE retrotransposons but does not contribute to the eye phenotype. Depletion of H3K36me2 methyltransferase ash1 in H3.3K27M, and of PRC2 component E(z) in H3.3K36M, restores the expression of eye developmental genes and normal eye growth, showing that redistribution of antagonistic marks contributes to K-to-M pathogenesis. Our results implicate a novel function for H3K36me2 and showcase convergent downstream effects of oncohistones that target opposing epigenetic marks.


Assuntos
Cromatina/química , Elementos de DNA Transponíveis , Histonas/química , Histonas/genética , Discos Imaginais/metabolismo , Mutação , Animais , Animais Geneticamente Modificados , Centrômero/ultraestrutura , Imunoprecipitação da Cromatina , Biologia Computacional/métodos , Metilação de DNA , Drosophila melanogaster , Epigênese Genética , Humanos , Lisina/química , Metionina/química , Camundongos , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Fenótipo , RNA-Seq
12.
Cell ; 154(2): 403-15, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23870128

RESUMO

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.


Assuntos
Autofagia , Metionina/metabolismo , Proteína Fosfatase 2/metabolismo , S-Adenosilmetionina/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metilação , Proteínas Metiltransferases/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/metabolismo
13.
Cell ; 153(1): 228-39, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23540700

RESUMO

The biguanide drug metformin is widely prescribed to treat type 2 diabetes and metabolic syndrome, but its mode of action remains uncertain. Metformin also increases lifespan in Caenorhabditis elegans cocultured with Escherichia coli. This bacterium exerts complex nutritional and pathogenic effects on its nematode predator/host that impact health and aging. We report that metformin increases lifespan by altering microbial folate and methionine metabolism. Alterations in metformin-induced longevity by mutation of worm methionine synthase (metr-1) and S-adenosylmethionine synthase (sams-1) imply metformin-induced methionine restriction in the host, consistent with action of this drug as a dietary restriction mimetic. Metformin increases or decreases worm lifespan, depending on E. coli strain metformin sensitivity and glucose concentration. In mammals, the intestinal microbiome influences host metabolism, including development of metabolic disease. Thus, metformin-induced alteration of microbial metabolism could contribute to therapeutic efficacy-and also to its side effects, which include folate deficiency and gastrointestinal upset.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/microbiologia , Ácido Fólico/metabolismo , Hipoglicemiantes/farmacologia , Longevidade/efeitos dos fármacos , Metformina/farmacologia , Metionina/metabolismo , Adenilato Quinase/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Biguanidas/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Restrição Calórica , Proteínas de Ligação a DNA/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Escherichia coli/metabolismo , Humanos , Hipoglicemiantes/metabolismo , Metagenoma , Metformina/metabolismo , Fatores de Transcrição/metabolismo
14.
Nature ; 601(7892): 245-251, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34912119

RESUMO

Pattern-triggered immunity (PTI) and effector-triggered immunity (ETI) in plants enable them to respond to pathogens by activating the production of defence metabolites that orchestrate immune responses1-4. How the production of defence metabolites is promoted by immune receptors and coordinated with broad-spectrum resistance remains elusive. Here we identify the deubiquitinase PICI1 as an immunity hub for PTI and ETI in rice (Oryza sativa). PICI1 deubiquitinates and stabilizes methionine synthetases to activate methionine-mediated immunity principally through biosynthesis of the phytohormone ethylene. PICI1 is targeted for degradation by blast fungal effectors, including AvrPi9, to dampen PTI. Nucleotide-binding domain, leucine-rich-repeat-containing receptors (NLRs) in the plant immune system, such as PigmR, protect PICI1 from effector-mediated degradation to reboot the methionine-ethylene cascade. Natural variation in the PICI1 gene contributes to divergence in basal blast resistance between the rice subspecies indica and japonica. Thus, NLRs govern an arms race with effectors, using a competitive mode that hinges on a critical defence metabolic pathway to synchronize PTI with ETI and ensure broad-spectrum resistance.


Assuntos
Oryza , Doenças das Plantas , Metionina , Oryza/genética , Oryza/microbiologia , Doenças das Plantas/microbiologia , Imunidade Vegetal/genética , Plantas , Transdução de Sinais/genética
15.
Mol Cell ; 78(2): 210-223.e8, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32208170

RESUMO

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.


Assuntos
Metilação de DNA/genética , Heterocromatina/genética , Metaboloma/genética , S-Adenosilmetionina/metabolismo , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Citoplasma/genética , Citoplasma/metabolismo , Epigênese Genética/genética , Regulação da Expressão Gênica/genética , Células HCT116 , Heterocromatina/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Metionina/genética , Camundongos , Processamento de Proteína Pós-Traducional/genética , Proteômica/métodos
16.
EMBO J ; 42(17): e114879, 2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37519235

RESUMO

The natural variation in mammalian longevity and its underlying mechanisms remain an active area of aging research. In the latest issue of The EMBO Journal, Liu et al (2023) analyze gene expression levels in 103 mammalian species across three tissues, revealing tissue-specific associations between gene expression patterns and longevity. Remarkably, the study suggests that methionine restriction, a strategy shown to increase lifespan, may extend beyond artificial interventions and is similarly employed by natural selection.


Assuntos
Longevidade , Metionina , Animais , Longevidade/genética , Metionina/genética , Metionina/metabolismo , Mamíferos/genética , Expressão Gênica , Envelhecimento/genética , Envelhecimento/metabolismo
17.
EMBO J ; 42(22): e114093, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37902464

RESUMO

Owing to their capability to disrupt the oxidative protein folding environment in the endoplasmic reticulum (ER), thiol antioxidants, such as dithiothreitol (DTT), are used as ER-specific stressors. We recently showed that thiol antioxidants modulate the methionine-homocysteine cycle by upregulating an S-adenosylmethionine-dependent methyltransferase, rips-1, in Caenorhabditis elegans. However, the changes in cellular physiology induced by thiol stress that modulate the methionine-homocysteine cycle remain uncharacterized. Here, using forward genetic screens in C. elegans, we discover that thiol stress enhances rips-1 expression via the hypoxia response pathway. We demonstrate that thiol stress activates the hypoxia response pathway. The activation of the hypoxia response pathway by thiol stress is conserved in human cells. The hypoxia response pathway enhances thiol toxicity via rips-1 expression and confers protection against thiol toxicity via rips-1-independent mechanisms. Finally, we show that DTT might activate the hypoxia response pathway by producing hydrogen sulfide. Our studies reveal an intriguing interaction between thiol-mediated reductive stress and the hypoxia response pathway and challenge the current model that thiol antioxidant DTT disrupts only the ER milieu in the cell.


Assuntos
Caenorhabditis elegans , Retículo Endoplasmático , Animais , Humanos , Caenorhabditis elegans/genética , Retículo Endoplasmático/metabolismo , Antioxidantes , Hipóxia/genética , Hipóxia/metabolismo , Homocisteína/metabolismo , Metionina/metabolismo , Estresse do Retículo Endoplasmático
18.
Proc Natl Acad Sci U S A ; 121(26): e2320835121, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38900797

RESUMO

Upper aerodigestive squamous cell carcinoma (UASCC) is a common and aggressive malignancy with few effective therapeutic options. Here, we investigate amino acid metabolism in this cancer, surprisingly noting that UASCC exhibits the highest methionine level across all human cancers, driven by its transporter LAT1. We show that LAT1 is also expressed at the highest level in UASCC, transcriptionally activated by UASCC-specific promoter and enhancers, which are directly coregulated by SCC master regulators TP63/KLF5/SREBF1. Unexpectedly, unbiased bioinformatic screen identifies EZH2 as the most significant target downstream of the LAT1-methionine pathway, directly linking methionine metabolism to epigenomic reprogramming. Importantly, this cascade is indispensable for the survival and proliferation of UASCC patient-derived tumor organoids. In addition, LAT1 expression is closely associated with cellular sensitivity to inhibition of the LAT1-methionine-EZH2 axis. Notably, this unique LAT1-methionine-EZH2 cascade can be targeted effectively by either pharmacological approaches or dietary intervention in vivo. In summary, this work maps a unique mechanistic cross talk between epigenomic reprogramming with methionine metabolism, establishes its biological significance in the biology of UASCC, and identifies a unique tumor-specific vulnerability which can be exploited both pharmacologically and dietarily.


Assuntos
Carcinoma de Células Escamosas , Regulação Neoplásica da Expressão Gênica , Transportador 1 de Aminoácidos Neutros Grandes , Metionina , Metionina/metabolismo , Humanos , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Linhagem Celular Tumoral , Epigênese Genética , Epigenômica/métodos , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Camundongos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Animais , Proliferação de Células , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Reprogramação Celular/genética
19.
Development ; 150(21)2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37846748

RESUMO

Histone modifications are associated with regulation of gene expression that controls a vast array of biological processes. Often, these associations are drawn by correlating the genomic location of a particular histone modification with gene expression or phenotype; however, establishing a causal relationship between histone marks and biological processes remains challenging. Consequently, there is a strong need for experimental approaches to directly manipulate histone modifications. A class of mutations on the N-terminal tail of histone H3, lysine-to-methionine (K-to-M) mutations, was identified as dominant-negative inhibitors of histone methylation at their respective and specific residues. The dominant-negative nature of K-to-M mutants makes them a valuable tool for studying the function of specific methylation marks on histone H3. Here, we review recent applications of K-to-M mutations to understand the role of histone methylation during development and homeostasis. We highlight important advantages and limitations that require consideration when using K-to-M mutants, particularly in a developmental context.


Assuntos
Cromatina , Histonas , Histonas/metabolismo , Cromatina/genética , Metilação , Mutação/genética , Metionina/genética , Metionina/metabolismo
20.
Development ; 150(20)2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36975381

RESUMO

Methionine is important for intestinal development and homeostasis in various organisms. However, the underlying mechanisms are poorly understood. Here, we demonstrate that the methionine adenosyltransferase gene Mat2a is essential for intestinal development and that the metabolite S-adenosyl-L-methionine (SAM) plays an important role in intestinal homeostasis. Intestinal epithelial cell (IEC)-specific knockout of Mat2a exhibits impaired intestinal development and neonatal lethality. Mat2a deletion in the adult intestine reduces cell proliferation and triggers IEC apoptosis, leading to severe intestinal epithelial atrophy and intestinal inflammation. Mechanistically, we reveal that SAM maintains the integrity of differentiated epithelium and protects IECs from apoptosis by suppressing the expression of caspases 3 and 8 and their activation. SAM supplementation improves the defective intestinal epithelium and reduces inflammatory infiltration sequentially. In conclusion, our study demonstrates that methionine metabolism and its intermediate metabolite SAM play essential roles in intestinal development and homeostasis in mice.


Assuntos
Metionina Adenosiltransferase , S-Adenosilmetionina , Camundongos , Animais , S-Adenosilmetionina/metabolismo , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Mucosa Intestinal/metabolismo , Metionina , Suplementos Nutricionais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA