Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.191
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 180(1): 135-149.e14, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31883797

RESUMEN

Autophagy is a conserved catabolic homeostasis process central for cellular and organismal health. During autophagy, small single-membrane phagophores rapidly expand into large double-membrane autophagosomes to encapsulate diverse cargoes for degradation. It is thought that autophagic membranes are mainly derived from preformed organelle membranes. Instead, here we delineate a pathway that expands the phagophore membrane by localized phospholipid synthesis. Specifically, we find that the conserved acyl-CoA synthetase Faa1 accumulates on nucleated phagophores and locally activates fatty acids (FAs) required for phagophore elongation and autophagy. Strikingly, using isotopic FA tracing, we directly show that Faa1 channels activated FAs into the synthesis of phospholipids and promotes their assembly into autophagic membranes. Indeed, the first committed steps of de novo phospholipid synthesis at the ER, which forms stable contacts with nascent autophagosomes, are essential for autophagy. Together, our work illuminates how cells spatially tune synthesis and flux of phospholipids for autophagosome biogenesis during autophagy.


Asunto(s)
Autofagia/fisiología , Ácidos Grasos/metabolismo , Fagosomas/metabolismo , Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Membrana Celular/metabolismo , Coenzima A Ligasas/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Fagosomas/fisiología , Fosfolípidos/biosíntesis , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
2.
Annu Rev Biochem ; 80: 859-83, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21275641

RESUMEN

The yeast Saccharomyces cerevisiae, with its full complement of organelles, synthesizes membrane phospholipids by pathways that are generally common to those found in higher eukaryotes. Phospholipid synthesis in yeast is regulated in response to a variety of growth conditions (e.g., inositol supplementation, zinc depletion, and growth stage) by a coordination of genetic (e.g., transcriptional activation and repression) and biochemical (e.g., activity modulation and localization) mechanisms. Phosphatidate (PA), whose cellular levels are controlled by the activities of key phospholipid synthesis enzymes, plays a central role in the transcriptional regulation of phospholipid synthesis genes. In addition to the regulation of gene expression, phosphorylation of key phospholipid synthesis catalytic and regulatory proteins controls the metabolism of phospholipid precursors and products.


Asunto(s)
Fosfolípidos/biosíntesis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Regulación Fúngica de la Expresión Génica , Inositol/metabolismo , Redes y Vías Metabólicas , Estructura Molecular , Fosfolípidos/química , Fosforilación , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Zinc/metabolismo
3.
J Biol Chem ; 299(12): 105417, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37918807

RESUMEN

In Saccharomyces cerevisiae, the transcriptional repressor Opi1 regulates the expression of genes involved in phospholipid synthesis responding to the abundance of the phospholipid precursor phosphatidic acid at the endoplasmic reticulum. We report here the identification of the conserved leucine zipper (LZ) domain of Opi1 as a hot spot for gain of function mutations and the characterization of the strongest variant identified, Opi1N150D. LZ modeling posits asparagine 150 embedded on the hydrophobic surface of the zipper and specifying dynamic parallel homodimerization by allowing electrostatic bonding across the hydrophobic dimerization interface. Opi1 variants carrying any of the other three ionic residues at amino acid 150 were also repressing. Genetic analyses showed that Opi1N150D variant is dominant, and its phenotype is attenuated when loss of function mutations identified in the other two conserved domains are present in cis. We build on the notion that membrane binding facilitates LZ dimerization to antagonize an intramolecular interaction of the zipper necessary for repression. Dissecting Opi1 protein in three polypeptides containing each conserved region, we performed in vitro analyses to explore interdomain interactions. An Opi11-190 probe interacted with Opi1291-404, the C terminus that bears the activator interacting domain (AID). LZ or AID loss of function mutations attenuated the interaction of the probes but was unaffected by the N150D mutation. We propose a model for Opi1 signal transduction whereby synergy between membrane-binding events and LZ dimerization antagonizes intramolecular LZ-AID interaction and transcriptional repression.


Asunto(s)
Leucina Zippers , Fosfolípidos , Proteínas Represoras , Proteínas de Saccharomyces cerevisiae , Fosfolípidos/biosíntesis , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Multimerización de Proteína
4.
PLoS Genet ; 17(12): e1009586, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34941903

RESUMEN

The cell envelope is essential for viability in all domains of life. It retains enzymes and substrates within a confined space while providing a protective barrier to the external environment. Destabilising the envelope of bacterial pathogens is a common strategy employed by antimicrobial treatment. However, even in one of the best studied organisms, Escherichia coli, there remain gaps in our understanding of how the synthesis of the successive layers of the cell envelope are coordinated during growth and cell division. Here, we used a whole-genome phenotypic screen to identify mutants with a defective cell envelope. We report that loss of yhcB, a conserved gene of unknown function, results in loss of envelope stability, increased cell permeability and dysregulated control of cell size. Using whole genome transposon mutagenesis strategies, we report the comprehensive genetic interaction network of yhcB, revealing all genes with a synthetic negative and a synthetic positive relationship. These genes include those previously reported to have a role in cell envelope biogenesis. Surprisingly, we identified genes previously annotated as essential that became non-essential in a ΔyhcB background. Subsequent analyses suggest that YhcB functions at the junction of several envelope biosynthetic pathways coordinating the spatiotemporal growth of the cell, highlighting YhcB as an as yet unexplored antimicrobial target.


Asunto(s)
Pared Celular/genética , Proteínas de Escherichia coli/genética , Lipopolisacáridos/genética , Oxidorreductasas/genética , Peptidoglicano/genética , División Celular/genética , Membrana Celular/genética , Membrana Celular/microbiología , Pared Celular/microbiología , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Lipopolisacáridos/biosíntesis , Mutagénesis , Fosfolípidos/biosíntesis , Fosfolípidos/genética
5.
J Hepatol ; 78(4): 820-835, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36681162

RESUMEN

BACKGROUND & AIMS: Hepatocyte growth and proliferation depends on membrane phospholipid biosynthesis. Short-chain fatty acids (SCFAs) generated by bacterial fermentation, delivered through the gut-liver axis, significantly contribute to lipid biosynthesis. We therefore hypothesized that dysbiotic insults like antibiotic treatment not only affect gut microbiota, but also impair hepatic lipid synthesis and liver regeneration. METHODS: Stable isotope labeling and 70% partial hepatectomy (PHx) was carried out in C57Bl/6J wild-type mice, in mice treated with broad-spectrum antibiotics, in germ-free mice and mice colonized with minimal microbiota. The microbiome was analyzed by 16S rRNA gene sequencing and microbial culture. Gut content, liver, blood and primary hepatocyte organoids were tested by mass spectrometry-based lipidomics, quantitative reverse-transcription PCR (qRT-PCR), immunoblot and immunohistochemistry for expression of proliferative and lipogenic markers. Matched biopsies from hyperplastic and hypoplastic liver tissue of patients subjected to surgical intervention to induce hyperplasia were analyzed by qRT-PCR for lipogenic enzymes. RESULTS: Three days of antibiotic treatment induced persistent dysbiosis with significantly decreased beta-diversity and richness, but a massive increase of Proteobacteria, accompanied by decreased colonic SCFAs. After PHx, antibiotic-treated mice showed delayed liver regeneration, increased mortality, impaired hepatocyte proliferation and decreased hepatic phospholipid synthesis. Expression of the lipogenic enzyme SCD1 was upregulated after PHx but delayed by antibiotic treatment. Germ-free mice essentially recapitulated the phenotype of antibiotic treatment. Phospholipid biosynthesis, hepatocyte proliferation, liver regeneration and survival were rescued in gnotobiotic mice colonized with a minimal SCFA-producing microbial community. SCFAs induced the growth of murine hepatocyte organoids and hepatic SCD1 expression in mice. Further, SCD1 was required for proliferation of human hepatoma cells and was associated with liver regeneration in human patients. CONCLUSION: Gut microbiota are pivotal for hepatic membrane phospholipid biosynthesis and liver regeneration. IMPACT AND IMPLICATIONS: Gut microbiota affect hepatic lipid metabolism through the gut-liver axis, but the underlying mechanisms are poorly understood. Perturbations of the gut microbiome, e.g. by antibiotics, impair the production of bacterial metabolites, which normally serve as building blocks for membrane lipids in liver cells. As a consequence, liver regeneration and survival after liver surgery is severely impaired. Even though this study is preclinical, its results might allow physicians in the future to improve patient outcomes after liver surgery, by modulation of gut microbiota or their metabolites.


Asunto(s)
Membrana Celular , Microbioma Gastrointestinal , Hepatocitos , Regeneración Hepática , Fosfolípidos , Animales , Humanos , Ratones , Antibacterianos/farmacología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Hiperplasia/metabolismo , Hiperplasia/patología , Hígado/patología , Regeneración Hepática/fisiología , Ratones Endogámicos C57BL , Fosfolípidos/biosíntesis , Fosfolípidos/metabolismo , ARN Ribosómico 16S , Hepatocitos/metabolismo , Membrana Celular/metabolismo
6.
Curr Genet ; 69(4-6): 289-300, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37947853

RESUMEN

Binding of general transcription factors TFIID and TFIIA to basal promoters is rate-limiting for transcriptional initiation of eukaryotic protein-coding genes. Consequently, activator proteins interacting with subunits of TFIID and/or TFIIA can drastically increase the rate of initiation events. Yeast transcriptional activator Ino2 interacts with several Taf subunits of TFIID, among them the multifunctional Taf1 protein. In contrast to mammalian Taf1, yeast Taf1 lacks bromodomains which are instead encoded by separate proteins Bdf1 and Bdf2. In this work, we show that Bdf1 not only binds to acetylated histone H4 but can also be recruited by Ino2 and unrelated activators such as Gal4, Rap1, Leu3 and Flo8. An activator-binding domain was mapped in the N-terminus of Bdf1. Subunits Toa1 and Toa2 of yeast TFIIA directly contact sequences of basal promoters and TFIID subunit TBP but may also mediate the influence of activators. Indeed, Ino2 efficiently binds to two separate structural domains of Toa1, specifically with its N-terminal four-helix bundle structure required for dimerization with Toa2 and its C-terminal ß-barrel domain contacting TBP and sequences of the TATA element. These findings complete the functional analysis of yeast general transcription factors Bdf1 and Toa1 and identify them as targets of activator proteins.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas que Contienen Bromodominio , Fosfolípidos , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Factor de Transcripción TFIIA , Factores de Transcripción , Fosfolípidos/biosíntesis , Fosfolípidos/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Factor de Transcripción TFIIA/genética , Factor de Transcripción TFIIA/metabolismo , Factor de Transcripción TFIID/genética , Factor de Transcripción TFIID/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas que Contienen Bromodominio/genética , Proteínas que Contienen Bromodominio/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(44): 27627-27636, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33087565

RESUMEN

Dengue virus (DENV) subdues cell membranes for its cellular cycle by reconfiguring phospholipids in humans and mosquitoes. Here, we determined how and why DENV reconfigures phospholipids in the mosquito vector. By inhibiting and activating the de novo phospholipid biosynthesis, we demonstrated the antiviral impact of de novo-produced phospholipids. In line with the virus hijacking lipids for its benefit, metabolomics analyses indicated that DENV actively inhibited the de novo phospholipid pathway and instead triggered phospholipid remodeling. We demonstrated the early induction of remodeling during infection by using isotope tracing in mosquito cells. We then confirmed in mosquitoes the antiviral impact of de novo phospholipids by supplementing infectious blood meals with a de novo phospholipid precursor. Eventually, we determined that phospholipid reconfiguration was required for viral genome replication but not for the other steps of the virus cellular cycle. Overall, we now propose that DENV reconfigures phospholipids through the remodeling cycle to modify the endomembrane and facilitate formation of the replication complex. Furthermore, our study identified de novo phospholipid precursor as a blood determinant of DENV human-to-mosquito transmission.


Asunto(s)
Aedes/virología , Virus del Dengue/fisiología , Dengue/transmisión , Mosquitos Vectores/virología , Fosfolípidos/biosíntesis , Aedes/enzimología , Animales , Línea Celular , Membrana Celular/metabolismo , Dengue/prevención & control , Dengue/virología , Virus del Dengue/patogenicidad , Genoma Viral , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Metabolismo de los Lípidos/genética , Redes y Vías Metabólicas/genética , Metabolómica , Mosquitos Vectores/enzimología , Interferencia de ARN , ARN Viral/metabolismo , Replicación Viral
8.
PLoS Genet ; 16(11): e1009192, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33227003

RESUMEN

During infection, cellular resources are allocated toward the metabolically-demanding processes of synthesizing and secreting effector proteins that neutralize and kill invading pathogens. In Drosophila, these effectors are antimicrobial peptides (AMPs) that are produced in the fat body, an organ that also serves as a major lipid storage depot. Here we asked how activation of Toll signaling in the larval fat body perturbs lipid homeostasis to understand how cells meet the metabolic demands of the immune response. We find that genetic or physiological activation of fat body Toll signaling leads to a tissue-autonomous reduction in triglyceride storage that is paralleled by decreased transcript levels of the DGAT homolog midway, which carries out the final step of triglyceride synthesis. In contrast, Kennedy pathway enzymes that synthesize membrane phospholipids are induced. Mass spectrometry analysis revealed elevated levels of major phosphatidylcholine and phosphatidylethanolamine species in fat bodies with active Toll signaling. The ER stress mediator Xbp1 contributed to the Toll-dependent induction of Kennedy pathway enzymes, which was blunted by deleting AMP genes, thereby reducing secretory demand elicited by Toll activation. Consistent with ER stress induction, ER volume is expanded in fat body cells with active Toll signaling, as determined by transmission electron microscopy. A major functional consequence of reduced Kennedy pathway induction is an impaired immune response to bacterial infection. Our results establish that Toll signaling induces a shift in anabolic lipid metabolism to favor phospholipid synthesis and ER expansion that may serve the immediate demand for AMP synthesis and secretion but with the long-term consequence of insufficient nutrient storage.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Infecciones por Bacterias Grampositivas/inmunología , Inmunidad Innata , Metabolismo de los Lípidos/inmunología , Animales , Animales Modificados Genéticamente , Péptidos Catiónicos Antimicrobianos/genética , Citidililtransferasa de Colina-Fosfato/genética , Citidililtransferasa de Colina-Fosfato/metabolismo , Proteínas de Unión al ADN/metabolismo , Diacilglicerol O-Acetiltransferasa/metabolismo , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/inmunología , Enterococcus faecalis/inmunología , Cuerpo Adiposo/enzimología , Cuerpo Adiposo/inmunología , Femenino , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Larva/enzimología , Larva/inmunología , Metabolismo de los Lípidos/genética , Masculino , Fosfolípidos/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo , Triglicéridos/metabolismo
9.
J Lipid Res ; 62: 100100, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34331935

RESUMEN

Choline phospholipids (PLs) such as phosphatidylcholine (PC) and 1-alkyl-2-acyl-sn-glycerophosphocholine are important components for cell membranes and also serve as a source of several lipid mediators. These lipids are biosynthesized in mammals in the final step of the CDP-choline pathway by the choline phosphotransferases choline phosphotransferase 1 (CPT1) and choline/ethanolamine phosphotransferase 1 (CEPT1). However, the contributions of these enzymes to the de novo biosynthesis of lipids remain unknown. Here, we established and characterized CPT1- and CEPT1-deficient human embryonic kidney 293 cells. Immunohistochemical analyses revealed that CPT1 localizes to the trans-Golgi network and CEPT1 to the endoplasmic reticulum. Enzyme assays and metabolic labeling with radiolabeled choline demonstrated that loss of CEPT1 dramatically decreases choline PL biosynthesis. Quantitative PCR and reintroduction of CPT1 and CEPT1 revealed that the specific activity of CEPT1 was much higher than that of CPT1. LC-MS/MS analysis of newly synthesized lipid molecular species from deuterium-labeled choline also showed that these enzymes have similar preference for the synthesis of PC molecular species, but that CPT1 had higher preference for 1-alkyl-2-acyl-sn-glycerophosphocholine with PUFA than did CEPT1. The endogenous level of PC was not reduced by the loss of these enzymes. However, several 1-alkyl-2-acyl-sn-glycerophosphocholine molecular species were reduced in CPT1-deficient cells and increased in CEPT1-deficient cells when cultured in 0.1% FBS medium. These results suggest that CEPT1 accounts for most choline PL biosynthesis activity, and that both enzymes are responsible for the production of different lipid molecular species in distinct organelles.


Asunto(s)
Colina/biosíntesis , Diacilglicerol Colinafosfotransferasa/metabolismo , Fosfolípidos/biosíntesis , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Células Cultivadas , Células HEK293 , Humanos
10.
J Biol Chem ; 295(7): 2136-2147, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31796629

RESUMEN

PlsX plays a central role in the coordination of fatty acid and phospholipid biosynthesis in Gram-positive bacteria. PlsX is a peripheral membrane acyltransferase that catalyzes the conversion of acyl-ACP to acyl-phosphate, which is in turn utilized by the polytopic membrane acyltransferase PlsY on the pathway of bacterial phospholipid biosynthesis. We have recently studied the interaction between PlsX and membrane phospholipids in vivo and in vitro, and observed that membrane association is necessary for the efficient transfer of acyl-phosphate to PlsY. However, understanding the molecular basis of such a channeling mechanism remains a major challenge. Here, we disentangle the binding and insertion events of the enzyme to the membrane, and the subsequent catalysis. We show that PlsX membrane binding is a process mostly mediated by phospholipid charge, whereas fatty acid saturation and membrane fluidity remarkably influence the membrane insertion step. Strikingly, the PlsXL254E mutant, whose biological functionality was severely compromised in vivo but remains catalytically active in vitro, was able to superficially bind to phospholipid vesicles, nevertheless, it loses the insertion capacity, strongly supporting the importance of membrane insertion in acyl-phosphate delivery. We propose a mechanism in which membrane fluidity governs the insertion of PlsX and thus regulates the biosynthesis of phospholipids in Gram-positive bacteria. This model may be operational in other peripheral membrane proteins with an unprecedented impact in drug discovery/development strategies.


Asunto(s)
Proteínas Bacterianas/genética , Bacterias Grampositivas/genética , Fluidez de la Membrana/genética , Fosfolípidos/biosíntesis , Bacillus subtilis/genética , Enterococcus faecalis/genética , Escherichia coli/genética , Fosfatos/metabolismo , Fosfolípidos/genética
11.
J Biol Chem ; 295(7): 2148-2159, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31919098

RESUMEN

PlsX is the first enzyme in the pathway that produces phosphatidic acid in Gram-positive bacteria. It makes acylphosphate from acyl-acyl carrier protein (acyl-ACP) and is also involved in coordinating phospholipid and fatty acid biosyntheses. PlsX is a peripheral membrane enzyme in Bacillus subtilis, but how it associates with the membrane remains largely unknown. In the present study, using fluorescence microscopy, liposome sedimentation, differential scanning calorimetry, and acyltransferase assays, we determined that PlsX binds directly to lipid bilayers and identified its membrane anchoring moiety, consisting of a hydrophobic loop located at the tip of two amphipathic dimerization helices. To establish the role of the membrane association of PlsX in acylphosphate synthesis and in the flux through the phosphatidic acid pathway, we then created mutations and gene fusions that prevent PlsX's interaction with the membrane. Interestingly, phospholipid synthesis was severely hampered in cells in which PlsX was detached from the membrane, and results from metabolic labeling indicated that these cells accumulated free fatty acids. Because the same mutations did not affect PlsX transacylase activity, we conclude that membrane association is required for the proper delivery of PlsX's product to PlsY, the next enzyme in the phosphatidic acid pathway. We conclude that PlsX plays a dual role in phospholipid synthesis, acting both as a catalyst and as a chaperone protein that mediates substrate channeling into the pathway.


Asunto(s)
Proteínas Bacterianas/genética , Redes y Vías Metabólicas/genética , Ácidos Fosfatidicos/metabolismo , Fosfolípidos/biosíntesis , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Catálisis , Escherichia coli/genética , Escherichia coli/metabolismo , Ácidos Grasos/metabolismo , Lipogénesis/genética , Ácidos Fosfatidicos/genética , Fosfolípidos/genética
12.
Plant J ; 103(4): 1351-1371, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32412123

RESUMEN

Symbiotic rhizobia-legume interactions are energy-demanding processes, and the carbon supply from host cells that is critically required for nodulation and nitrogen fixation is not fully understood. Investigation of the lipidomic and carbohydrate profiles with the transcriptome of developing nodules revealed highly activated glycolysis, fatty acid (FA), 2-monoacylglycerol (2-MAG), and membrane lipid biosynthesis and transport during nodule development. RNA-sequence profiling of metabolic genes in roots and developing nodules highlighted the enhanced expression of genes involved in the biosynthesis and transport of FAs, membrane lipids, and 2-MAG in rhizobia-soybean symbioses via the RAML-WRI-FatM-GPAT-STRL pathway, which is similar to that in legume-arbuscular mycorrhizal fungi symbiosis. The essential roles of the metabolic pathway during soybean nodulation were further supported by analysis of transgenic hairy roots overexpressing soybean GmWRI1b-OE and GmLEC2a-OE. GmLEC2a-OE hairy roots produced fewer nodules, in contrast to GmWRI1b-OE hairy roots. GmLEC2a-OE hairy roots displayed different or even opposite expression patterns of the genes involved in glycolysis and the synthesis of FAs, 2-MAG, TAG, and membrane lipids compared to GmWRI1b-OE hairy roots. Glycolysis, FA and membrane lipid biosynthesis were repressed in GmLEC2a-OE but increased in GmWRI1b-OE hairy roots, which may account for the reduced nodulation in GmLEC2a-OE hairy roots but increased nodulation in GmWRI1b-OE hairy roots. These data show that active FA, 2-MAG and membrane lipid biosynthesis are essential for nodulation and rhizobia-soybean symbioses. These data shed light on essential and complex lipid metabolism for soybean nodulation and nodule development, laying the foundation for the future detailed investigation of soybean nodulation.


Asunto(s)
Ácidos Grasos/biosíntesis , Glycine max/metabolismo , Glucólisis , Lípidos/biosíntesis , Nodulación de la Raíz de la Planta/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Glucólisis/fisiología , Lipidómica , Lípidos de la Membrana/biosíntesis , Fosfolípidos/biosíntesis , Raíces de Plantas/metabolismo , Nódulos de las Raíces de las Plantas/metabolismo , Glycine max/crecimiento & desarrollo
13.
J Am Chem Soc ; 143(23): 8533-8537, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33978402

RESUMEN

The de novo formation of lipid membranes from minimal reactive precursors is a major goal in synthetic cell research. In nature, the synthesis of membrane phospholipids is orchestrated by numerous enzymes, including fatty acid synthases and membrane-bound acyltransferases. However, these enzymatic pathways are difficult to fully reproduce in vitro. As such, the reconstitution of phospholipid membrane synthesis from simple metabolic building blocks remains a challenge. Here, we describe a chemoenzymatic strategy for lipid membrane generation that utilizes a soluble bacterial fatty acid synthase (cgFAS I) to synthesize palmitoyl-CoA in situ from acetyl-CoA and malonyl-CoA. The fatty acid derivative spontaneously reacts with a cysteine-modified lysophospholipid by native chemical ligation (NCL), affording a noncanonical amidophospholipid that self-assembles into micron-sized membrane-bound vesicles. To our knowledge, this is the first example of reconstituting phospholipid membrane formation directly from acetyl-CoA and malonyl-CoA precursors. Our results demonstrate that combining the specificity and efficiency of a type I fatty acid synthase with a highly selective bioconjugation reaction provides a biomimetic route for the de novo formation of membrane-bound vesicles.


Asunto(s)
Acido Graso Sintasa Tipo I/metabolismo , Fosfolípidos/biosíntesis , Acido Graso Sintasa Tipo I/química , Estructura Molecular , Fosfolípidos/química
14.
Mol Microbiol ; 114(4): 653-663, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32671874

RESUMEN

A key aspect in membrane biogenesis is the coordination of fatty acid to phospholipid synthesis rates. In most bacteria, PlsX is the first enzyme of the phosphatidic acid synthesis pathway, the common precursor of all phospholipids. Previously, we proposed that PlsX is a key regulatory point that synchronizes the fatty acid synthase II with phospholipid synthesis in Bacillus subtilis. However, understanding the basis of such coordination mechanism remained a challenge in Gram-positive bacteria. Here, we show that the inhibition of fatty acid and phospholipid synthesis caused by PlsX depletion leads to the accumulation of long-chain acyl-ACPs, the end products of the fatty acid synthase II. Hydrolysis of the acyl-ACP pool by heterologous expression of a cytosolic thioesterase relieves the inhibition of fatty acid synthesis, indicating that acyl-ACPs are feedback inhibitors of this metabolic route. Unexpectedly, inactivation of PlsX triggers a large increase of malonyl-CoA leading to induction of the fap regulon. This finding discards the hypothesis, proposed for B. subtilis and extended to other Gram-positive bacteria, that acyl-ACPs are feedback inhibitors of the acetyl-CoA carboxylase. Finally, we propose that the continuous production of malonyl-CoA during phospholipid synthesis inhibition provides an additional mechanism for fine-tuning the coupling between phospholipid and fatty acid production in bacteria with FapR regulation.


Asunto(s)
Bacillus subtilis/metabolismo , Ácidos Grasos/biosíntesis , Fosfolípidos/biosíntesis , Proteína Transportadora de Acilo/metabolismo , Proteínas Bacterianas/metabolismo , Ácidos Grasos/metabolismo , Lipogénesis , Fosfolípidos/metabolismo , Regulón
15.
PLoS Biol ; 16(2): e2005188, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29481577

RESUMEN

Lipids are essential components of biological membranes that present a wide diversity in eukaryotic cells. Recent impressive advances in lipid biochemistry and biophysics have enabled a refocus of our view of lipids as functional units for cellular activity. However, the gap between molecular and cellular processes remains to be bridged. Here, 2 papers meet the burden of proof that choline transporters participate in local lipid composition modifications at the trans-Golgi network, an intracellular compartment that serves as the main sorting station in the cell. Localization of choline transporters to this precise compartment could be a way for plant cells to quickly modify the membrane lipid composition and asymmetry during both the allocation of cargos and the recruitment of trafficking machineries into distinct subcellular pathways.


Asunto(s)
Lípidos de la Membrana/metabolismo , Acetilcolina/metabolismo , Animales , Transporte Biológico , Compartimento Celular , Colina/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Fosfolípidos/biosíntesis , Prueba de Estudio Conceptual , Red trans-Golgi/metabolismo
16.
J Inherit Metab Dis ; 44(4): 809-825, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33594685

RESUMEN

Over 80 human diseases have been attributed to defects in complex lipid metabolism. A majority of them have been reported recently in the setting of rapid advances in genomic technology and their increased use in clinical settings. Lipids are ubiquitous in human biology and play roles in many cellular and intercellular processes. While inborn errors in lipid metabolism can affect every organ system with many examples of genetic heterogeneity and pleiotropy, the clinical manifestations of many of these disorders can be explained based on the disruption of the metabolic pathway involved. In this review, we will discuss the physiological function of major pathways in complex lipid metabolism, including nonlysosomal sphingolipid metabolism, acylceramide metabolism, de novo phospholipid synthesis, phospholipid remodeling, phosphatidylinositol metabolism, mitochondrial cardiolipin synthesis and remodeling, and ether lipid metabolism as well as common clinical phenotypes associated with each.


Asunto(s)
Metabolismo de los Lípidos , Lípidos/química , Redes y Vías Metabólicas/fisiología , Cardiolipinas/biosíntesis , Cardiolipinas/química , Homeostasis , Humanos , Lípidos/biosíntesis , Mitocondrias/metabolismo , Mitocondrias/patología , Fenotipo , Fosfatidilinositoles/biosíntesis , Fosfatidilinositoles/química , Fosfolípidos/biosíntesis , Fosfolípidos/química , Esfingolípidos/biosíntesis , Esfingolípidos/química
17.
Biochem J ; 477(23): 4675-4688, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33211090

RESUMEN

Glioblastoma multiforme is the most aggressive type of tumor of the CNS with an overall survival rate of approximately one year. Since this rate has not changed significantly over the last 20 years, the development of new therapeutic strategies for the treatment of these tumors is peremptory. The over-expression of the proto-oncogene c-Fos has been observed in several CNS tumors including glioblastoma multiforme and is usually associated with a poor prognosis. Besides its genomic activity as an AP-1 transcription factor, this protein can also activate phospholipid synthesis by a direct interaction with key enzymes of their metabolic pathways. Given that the amino-terminal portion of c-Fos (c-Fos-NA: amino acids 1-138) associates to but does not activate phospholipid synthesizing enzymes, we evaluated if c-Fos-NA or some shorter derivatives are capable of acting as dominant-negative peptides of the activating capacity of c-Fos. The over-expression or the exogenous administration of c-Fos-NA to cultured T98G cells hampers the interaction between c-Fos and PI4K2A, an enzyme activated by c-Fos. Moreover, it was observed a decrease in tumor cell proliferation rates in vitro and a reduction in tumor growth in vivo when a U87-MG-generated xenograft on nude mice is intratumorally treated with recombinant c-Fos-NA. Importantly, a smaller peptide of 92 amino acids derived from c-Fos-NA retains the capacity to interfere with tumor proliferation in vitro and in vivo. Taken together, these results support the use of the N-terminal portion of c-Fos, or shorter derivatives as a novel therapeutic strategy for the treatment of glioblastoma multiforme.


Asunto(s)
Proliferación Celular , Glioblastoma/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Fosfolípidos/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Línea Celular Tumoral , Activación Enzimática , Glioblastoma/genética , Glioblastoma/patología , Humanos , Antígenos de Histocompatibilidad Menor/genética , Fosfolípidos/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-fos/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
18.
Int J Mol Sci ; 23(1)2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-35008618

RESUMEN

The outer membrane (OM) of Gram-negative bacteria, such as Escherichia coli, is essential for their viability. Lipopolysaccharide (LPS) constitutes the major component of OM, providing the permeability barrier, and a tight balance exists between LPS and phospholipids amounts as both of these essential components use a common metabolic precursor. Hence, checkpoints are in place, right from the regulation of the first committed step in LPS biosynthesis mediated by LpxC through its turnover by FtsH and HslUV proteases in coordination with LPS assembly factors LapB and LapC. After the synthesis of LPS on the inner leaflet of the inner membrane (IM), LPS is flipped by the IM-located essential ATP-dependent transporter to the periplasmic face of IM, where it is picked up by the LPS transport complex spanning all three components of the cell envelope for its delivery to OM. MsbA exerts its intrinsic hydrocarbon ruler function as another checkpoint to transport hexa-acylated LPS as compared to underacylated LPS. Additional checkpoints in LPS assembly are: LapB-assisted coupling of LPS synthesis and translocation; cardiolipin presence when LPS is underacylated; the recruitment of RfaH transcriptional factor ensuring the transcription of LPS core biosynthetic genes; and the regulated incorporation of non-stoichiometric modifications, controlled by the stress-responsive RpoE sigma factor, small RNAs and two-component systems.


Asunto(s)
Escherichia coli/metabolismo , Lipopolisacáridos/biosíntesis , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Lipopolisacáridos/química , Modelos Biológicos , Mutación/genética , Fosfolípidos/biosíntesis , Proteolisis
19.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34445706

RESUMEN

Here, we present the main features of human acid sphingomyelinase (ASM), its biosynthesis, processing and intracellular trafficking, its structure, its broad substrate specificity, and the proposed mode of action at the surface of the phospholipid substrate carrying intraendolysosomal luminal vesicles. In addition, we discuss the complex regulation of its phospholipid cleaving activity by membrane lipids and lipid-binding proteins. The majority of the literature implies that ASM hydrolyses solely sphingomyelin to generate ceramide and ignores its ability to degrade further substrates. Indeed, more than twenty different phospholipids are cleaved by ASM in vitro, including some minor but functionally important phospholipids such as the growth factor ceramide-1-phosphate and the unique lysosomal lysolipid bis(monoacylglycero)phosphate. The inherited ASM deficiency, Niemann-Pick disease type A and B, impairs mainly, but not only, cellular sphingomyelin catabolism, causing a progressive sphingomyelin accumulation, which furthermore triggers a secondary accumulation of lipids (cholesterol, glucosylceramide, GM2) by inhibiting their turnover in late endosomes and lysosomes. However, ASM appears to be involved in a variety of major cellular functions with a regulatory significance for an increasing number of metabolic disorders. The biochemical characteristics of ASM, their potential effect on cellular lipid turnover, as well as a potential impact on physiological processes will be discussed.


Asunto(s)
Fosfolípidos/biosíntesis , Esfingomielina Fosfodiesterasa/biosíntesis , Esfingomielina Fosfodiesterasa/metabolismo , Transporte Biológico , Ceramidas/metabolismo , Colesterol/metabolismo , Endosomas/metabolismo , Humanos , Lisosomas/metabolismo , Lípidos de la Membrana/metabolismo , Enfermedad de Niemann-Pick Tipo A/metabolismo , Fosfolípidos/metabolismo , Esfingomielina Fosfodiesterasa/fisiología , Esfingomielinas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Fosfolipasas de Tipo C/fisiología
20.
J Lipid Res ; 61(8): 1232-1243, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32540926

RESUMEN

PA phosphatase, encoded by PAH1 in the yeast Saccharomyces cerevisiae, catalyzes the Mg2+-dependent dephosphorylation of PA, producing DAG at the nuclear/ER membrane. This enzyme plays a major role in triacylglycerol synthesis and in the regulation of phospholipid synthesis. As an interfacial enzyme, PA phosphatase interacts with the membrane surface, binds its substrate, and catalyzes its reaction. The Triton X-100/PA-mixed micellar system has been utilized to examine the activity and regulation of yeast PA phosphatase. This system, however, does not resemble the in vivo environment of the membrane phospholipid bilayer. We developed an assay system that mimics the nuclear/ER membrane to assess PA phosphatase activity. PA was incorporated into unilamellar phospholipid vesicles (liposomes) composed of the major nuclear/ER membrane phospholipids, PC, PE, PI, and PS. We optimized this system to support enzyme-liposome interactions and to afford activity that is greater than that obtained with the aforementioned detergent system. Activity was regulated by phospholipid composition, whereas the enzyme's interaction with liposomes was insensitive to composition. Greater activity was attained with large (≥100 nm) versus small (50 nm) vesicles. The fatty-acyl moiety of PA had no effect on this activity. PA phosphatase activity was dependent on the bulk (hopping mode) and surface (scooting mode) concentrations of PA, suggesting a mechanism by which the enzyme operates along the nuclear/ER membrane in vivo.


Asunto(s)
Membrana Dobles de Lípidos/metabolismo , Fosfatidato Fosfatasa/metabolismo , Fosfolípidos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Fosfolípidos/biosíntesis , Saccharomyces cerevisiae/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA