Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
1.
Annu Rev Immunol ; 38: 289-313, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31986069

ABSTRACT

A striking change has happened in the field of immunology whereby specific metabolic processes have been shown to be a critical determinant of immune cell activation. Multiple immune receptor types rewire metabolic pathways as a key part of how they promote effector functions. Perhaps surprisingly for immunologists, the Krebs cycle has emerged as the central immunometabolic hub of the macrophage. During proinflammatory macrophage activation, there is an accumulation of the Krebs cycle intermediates succinate and citrate, and the Krebs cycle-derived metabolite itaconate. These metabolites have distinct nonmetabolic signaling roles that influence inflammatory gene expression. A key bioenergetic target for the Krebs cycle, the electron transport chain, also becomes altered, generating reactive oxygen species from Complexes I and III. Similarly, alternatively activated macrophages require α-ketoglutarate-dependent epigenetic reprogramming to elicit anti-inflammatory gene expression. In this review, we discuss these advances and speculate on the possibility of targeting these events therapeutically for inflammatory diseases.


Subject(s)
Citric Acid Cycle , Immunity , Macrophages/immunology , Macrophages/metabolism , Animals , Disease Susceptibility , Energy Metabolism , Humans , Immunomodulation , Macrophage Activation/immunology , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 119(14): e2024357119, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35353621

ABSTRACT

Prostate epithelial cells have the unique capacity to secrete large amounts of citrate, but the carbon sources and metabolic pathways that maintain this production are not well known. We mapped potential pathways for citrate carbons in the human prostate cancer metastasis cell lines LNCaP and VCaP, for which we first established that they secrete citrate (For LNCaP 5.6 ± 0.9 nmol/h per 106 cells). Using 13C-labeled substrates, we traced the incorporation of 13C into citrate by NMR of extracellular fluid. Our results provide direct evidence that glucose is a main carbon source for secreted citrate. We also demonstrate that carbons from supplied glutamine flow via oxidative Krebs cycle and reductive carboxylation routes to positions in secreted citrate but likely do not contribute to its net synthesis. The potential anaplerotic carbon sources aspartate and asparagine did not contribute to citrate carbons. We developed a quantitative metabolic model employing the 13C distribution in extracellular citrate after 13C glucose and pyruvate application to assess intracellular pathways of carbons for secreted citrate. From this model, it was estimated that in LNCaP about 21% of pyruvate entering the Krebs cycle is converted via pyruvate carboxylase as an anaplerotic route at a rate more than sufficient to compensate carbon loss of this cycle by citrate secretion. This model provides an estimation of the fraction of molecules, including citrate, leaving the Krebs cycle at every turn. The measured ratios of 13C atoms at different positions in extracellular citrate may serve as biomarkers for (malignant) epithelial cell metabolism.


Subject(s)
Biomarkers, Tumor , Citric Acid , Prostatic Neoplasms , Biomarkers, Tumor/metabolism , Carbon/metabolism , Carbon Isotopes , Citrates , Citric Acid/metabolism , Citric Acid Cycle , Glucose/metabolism , Humans , Magnetic Resonance Spectroscopy , Male , Prostatic Neoplasms/metabolism
3.
J Biol Chem ; 299(2): 102838, 2023 02.
Article in English | MEDLINE | ID: mdl-36581208

ABSTRACT

The tricarboxylic acid (TCA) cycle, otherwise known as the Krebs cycle, is a central metabolic pathway that performs the essential function of oxidizing nutrients to support cellular bioenergetics. More recently, it has become evident that TCA cycle behavior is dynamic, and products of the TCA cycle can be co-opted in cancer and other pathologic states. In this review, we revisit the TCA cycle, including its potential origins and the history of its discovery. We provide a detailed accounting of the requirements for sustained TCA cycle function and the critical regulatory nodes that can stimulate or constrain TCA cycle activity. We also discuss recent advances in our understanding of the flexibility of TCA cycle wiring and the increasingly appreciated heterogeneity in TCA cycle activity exhibited by mammalian cells. Deeper insight into how the TCA cycle can be differentially regulated and, consequently, configured in different contexts will shed light on how this pathway is primed to meet the requirements of distinct mammalian cell states.


Subject(s)
Citric Acid Cycle , Energy Metabolism , Animals , Citric Acid Cycle/physiology , Mammals
4.
J Biol Chem ; 299(6): 104761, 2023 06.
Article in English | MEDLINE | ID: mdl-37119852

ABSTRACT

Mitochondrial complex II is traditionally studied for its participation in two key respiratory processes: the electron transport chain and the Krebs cycle. There is now a rich body of literature explaining how complex II contributes to respiration. However, more recent research shows that not all of the pathologies associated with altered complex II activity clearly correlate with this respiratory role. Complex II activity has now been shown to be necessary for a range of biological processes peripherally related to respiration, including metabolic control, inflammation, and cell fate. Integration of findings from multiple types of studies suggests that complex II both participates in respiration and controls multiple succinate-dependent signal transduction pathways. Thus, the emerging view is that the true biological function of complex II is well beyond respiration. This review uses a semichronological approach to highlight major paradigm shifts that occurred over time. Special emphasis is given to the more recently identified functions of complex II and its subunits because these findings have infused new directions into an established field.


Subject(s)
Electron Transport Complex II , Succinate Dehydrogenase , Citric Acid Cycle , Respiration , Signal Transduction , Succinate Dehydrogenase/metabolism , Mitochondria , Electron Transport Complex II/metabolism
5.
J Neurosci Res ; 102(5): e25339, 2024 May.
Article in English | MEDLINE | ID: mdl-38741550

ABSTRACT

Diets rich in saturated fats are more detrimental to health than those containing mono- or unsaturated fats. Fatty acids are an important source of energy, but they also relay information regarding nutritional status to hypothalamic metabolic circuits and when in excess can be detrimental to these circuits. Astrocytes are the main site of central fatty acid ß-oxidation, and hypothalamic astrocytes participate in energy homeostasis, in part by modulating hormonal and nutritional signals reaching metabolic neurons, as well as in the inflammatory response to high-fat diets. Thus, we hypothesized that how hypothalamic astrocytes process-specific fatty acids participates in determining the differential metabolic response and that this is sex dependent as males and females respond differently to high-fat diets. Male and female primary hypothalamic astrocyte cultures were treated with oleic acid (OA) or palmitic acid (PA) for 24 h, and an untargeted metabolomics study was performed. A clear predictive model for PA exposure was obtained, while the metabolome after OA exposure was not different from controls. The observed modifications in metabolites, as well as the expression levels of key metabolic enzymes, indicate a reduction in the activity of the Krebs and glutamate/glutamine cycles in response to PA. In addition, there were specific differences between the response of astrocytes from male and female mice, as well as between hypothalamic and cerebral cortical astrocytes. Thus, the response of hypothalamic astrocytes to specific fatty acids could result in differential impacts on surrounding metabolic neurons and resulting in varied systemic metabolic outcomes.


Subject(s)
Astrocytes , Hypothalamus , Oleic Acid , Palmitic Acid , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Oleic Acid/pharmacology , Female , Palmitic Acid/pharmacology , Hypothalamus/metabolism , Hypothalamus/drug effects , Male , Mice , Mice, Inbred C57BL , Sex Characteristics , Cells, Cultured
6.
Clin Immunol ; 246: 109216, 2023 01.
Article in English | MEDLINE | ID: mdl-36572212

ABSTRACT

Macrophages are a diverse population of phagocytic immune cells involved in the host defense mechanisms and regulation of homeostasis. Usually, macrophages maintain healthy functioning at the cellular level, but external perturbation in their balanced functions can lead to acute and chronic disease conditions. By sensing the cues from the tissue microenvironment, these phagocytes adopt a plethora of phenotypes, such as inflammatory or M1 to anti-inflammatory (immunosuppressive) or M2 subtypes, to fulfill their spectral range of functions. The existing evidence in the literature supports that in macrophages, regulation of metabolic switches and metabolic adaptations are associated with their functional behaviors under various physiological and pathological conditions. Since these macrophages play a crucial role in many disorders, therefore it is necessary to understand their heterogeneity and metabolic reprogramming. Consequently, these macrophages have also emerged as a promising target for diseases in which their role is crucial in driving the disease pathology and outcome (e.g., Cancers). In this review, we discuss the recent findings that link many metabolites with macrophage functions and highlight how this metabolic reprogramming can improve our understanding of cellular malfunction in the macrophages during inflammatory disorders. A systematic analysis of the interconnecting crosstalk between metabolic pathways with macrophages should inform the selection of immunomodulatory therapies for inflammatory diseases.


Subject(s)
Inflammation , Macrophages , Humans , Phagocytes , Anti-Inflammatory Agents , Phenotype
7.
Toxicol Appl Pharmacol ; 465: 116451, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36894070

ABSTRACT

The toxic effects of alcohol consumption on population health are significant worldwide and the synergistic toxic effects of concurrent intake of Acetaminophen and alcohol is of clinical concern. The understanding of molecular mechanisms beneath such synergism and acute toxicity may be enhanced through assessing underlying metabolomics changes. The molecular toxic activities of the model hereby, is assessed though metabolomics profile with a view to identifying metabolomics targets which could aid in the management of drug-alcohol interactions. In vivo exposure of C57/BL6 mice to APAP (70 mg/kg), single dose of ethanol (6 g/kg of 40%) and APAP after alcohol consumption was employed. Plasma samples were prepared and subjected to biphasic extraction for complete LC-MS profiling, and tandem mass MS2 analysis. Among the detected ions, 174 ions had significant (VIP scores >1 and FDR <0.05) changes between groups and were selected as potential biomarkers and significant variables. The presented metabolomics approach highlighted several affected metabolic pathways, including nucleotide and amino acid metabolism; aminoacyl-tRNA biosynthesis as well as bioenergetics of TCA and Krebs cycle. The impact of APAP on the concurrent administration of alcohol showed great biological interactions in the vital ATP and amino acid producing processes. The metabolomics changes show distinct metabolites which are altered to alcohol-APAP consumption while presenting several unneglectable risks on the vitality of metabolites and cellular molecules which shall be concerned.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Mice , Animals , Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Liver , Metabolomics , Biomarkers , Amino Acids/metabolism , Alcohol Drinking/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Mice, Inbred C57BL
8.
Trends Biochem Sci ; 43(11): 847-849, 2018 11.
Article in English | MEDLINE | ID: mdl-29807701

ABSTRACT

Hans Kornberg wrote a paper entitled 'Krebs and his trinity of cycles' commenting that every school biology student knows of the Krebs cycle, but few know that Krebs discovered two other cycles. These are (i) the ornithine cycle (urea cycle), (ii) the citric acid cycle (tricarboxylic acid or TCA cycle), and (iii) the glyoxylate cycle that was described by Krebs and Kornberg. Ironically, Kornberg, codiscoverer of the 'glyoxylate cycle', overlooked a fourth Krebs cycle - (iv) the uric acid cycle.


Subject(s)
Citric Acid Cycle , Glyoxylates/metabolism , Ornithine/metabolism , Urea/metabolism , Uric Acid/metabolism , Humans
9.
J Biol Chem ; 297(1): 100839, 2021 07.
Article in English | MEDLINE | ID: mdl-34051232

ABSTRACT

Glucose-mediated signaling regulates the expression of a limited number of genes in human pancreatic ß-cells at the transcriptional level. However, it is unclear whether glucose plays a role in posttranscriptional RNA processing or translational control of gene expression. Here, we asked whether glucose affects posttranscriptional steps and regulates protein synthesis in human ß-cell lines. We first showed the involvement of the mTOR pathway in glucose-related signaling. We also used the surface sensing of translation technique, based on puromycin incorporation into newly translated proteins, to demonstrate that glucose treatment increased protein translation. Among the list of glucose-induced proteins, we identified the proconvertase PCSK1, an enzyme involved in the proteolytic conversion of proinsulin to insulin, whose translation was induced within minutes following glucose treatment. We finally performed global proteomic analysis by mass spectrometry to characterize newly translated proteins upon glucose treatment. We found enrichment in proteins involved in translation, glycolysis, TCA metabolism, and insulin secretion. Taken together, our study demonstrates that, although glucose minorly affects gene transcription in human ß-cells, it plays a major role at the translational level.


Subject(s)
Energy Metabolism/genetics , Glucose/pharmacology , Insulin Secretion/genetics , Insulin-Secreting Cells/metabolism , Protein Biosynthesis/genetics , Cell Line , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/metabolism , Energy Metabolism/drug effects , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Mitogen-Activated Protein Kinases/metabolism , Proprotein Convertase 1/metabolism , Protein Biosynthesis/drug effects , Puromycin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
10.
J Cell Biochem ; 123(4): 701-718, 2022 04.
Article in English | MEDLINE | ID: mdl-34931340

ABSTRACT

Acetylation of proteins seems a widespread process found in the three domains of life. Several studies have shown that besides histones, acetylation of lysine residues also occurs in non-nuclear proteins. Hence, it has been suggested that this covalent modification is a mechanism that might regulate diverse metabolic pathways by modulating enzyme activity, stability, and/or subcellular localization or interaction with other proteins. However, protein acetylation levels seem to have low correlation with modification of enzyme activity and pathway fluxes. In addition, the results obtained with mutant enzymes that presumably mimic acetylation have frequently been over-interpreted. Moreover, there is a generalized lack of rigorous enzyme kinetic analysis in parallel to acetylation level determinations. The purpose of this review is to analyze the current findings on the impact of acetylation on metabolic enzymes and its repercussion on metabolic pathways function/regulation.


Subject(s)
Metabolic Networks and Pathways , Protein Processing, Post-Translational , Acetylation , Histones , Kinetics
11.
Am J Hum Genet ; 105(4): 813-821, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31564436

ABSTRACT

Germline heterozygous PTEN mutations cause subsets of Cowden syndrome (CS) and Bannayan-Riley-Ruvalcaba syndrome (BRRS); these subsets are characterized by high risks of breast, thyroid, and other cancers and, in one subset, autism spectrum disorder (ASD). Up to 10% of individuals with PTENMUT CS, CS-like syndrome, or BRRS have germline SDHx (succinate dehydrogenase, mitochondrial complex II) variants, which modify cancer risk. PTEN contributes to metabolic reprogramming; this is a well-established role in a cancer context. Relatedly, SDH sits at the crossroad of the electron transport chain and tricarboxylic acid (TCA) cycle, two central bioenergetic pathways. Intriguingly, PTENMUT and SDHMUT individuals have reduced SDH catalytic activity, resulting in succinate accumulation; this indicates a common genotype-independent biochemical alteration. Here, we conducted a TCA targeted metabolomics study on 511 individuals with CS, CS-like syndrome, or BRRS with various genotypes (PTEN or SDHx, mutant or wild type [WT]) and phenotypes (cancer or ASD) and a series of 187 population controls. We found consistent TCA cycle metabolite alterations in cases with various genotypes and phenotypes compared to controls, and we found unique correlations of individual metabolites with particular genotype-phenotype combinations. Notably, increased isocitrate (p = 1.2 × 10-3), but reduced citrate (p = 5.0 × 10-4), were found to be associated with breast cancer in individuals with PTENMUT/SDHxWT. Conversely, increased lactate was associated with neurodevelopmental disorders regardless of genotype (p = 9.7 × 10-3); this finding was replicated in an independent validation series (n = 171) enriched for idiopathic ASD (PTENWT, p = 5.6 × 10-4). Importantly, we identified fumarate (p = 1.9 × 10-2) as a pertinent metabolite, distinguishing individuals who develop ASD from those who develop cancer. Our observations suggest that TCA cycle metabolite alterations are germane to the pathobiology of PTEN-related CS and BRRS, as well as genotype-independent ASD, with implications for potential biomarker and/or therapeutic value.


Subject(s)
Autistic Disorder/genetics , Citric Acid Cycle , Hamartoma Syndrome, Multiple/genetics , Neoplasms/genetics , Phenotype , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Young Adult
12.
Clin Genet ; 101(2): 260-264, 2022 02.
Article in English | MEDLINE | ID: mdl-34766628

ABSTRACT

Bi-allelic alterations in the MDH2 gene have recently been reported in three unrelated toddlers with early-onset severe encephalopathy. Here, we describe a new case of a child carrying novel variants in MDH2. This child presented with early-onset encephalocardiopathy requiring heart transplant and showed cerebellar ataxia and drug-responsive epilepsy; his family history was significant for multiple cancers, a feature often associated with monoallelic variants in MDH2. Functional studies in cultured skin fibroblasts from the proband showed reduced protein levels and impaired enzyme activity, further corroborating the genetic results. The relatively mild neurological presentation and severe cardiac manifestations requiring heart transplant distinguish this case from previous reports. This patient thus expands the spectrum of clinical features associated with MDH2 variants.


Subject(s)
Alleles , Genetic Association Studies , Genetic Predisposition to Disease , Malate Dehydrogenase/genetics , Mutation , Phenotype , Child , Child, Preschool , DNA Mutational Analysis , Genome, Mitochondrial , Humans , Infant , Magnetic Resonance Imaging , Neuroimaging , Exome Sequencing
13.
Mar Drugs ; 20(2)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35200644

ABSTRACT

Docosahexaenoic acid (DHA) is one of the most important long-chain polyunsaturated fatty acids (LC-PUFAs), with numerous health benefits. Crypthecodinium cohnii, a marine heterotrophic dinoflagellate, is successfully used for the industrial production of DHA because it can accumulate DHA at high concentrations within the cells. Glycerol is an interesting renewable substrate for DHA production since it is a by-product of biodiesel production and other industries, and is globally generated in large quantities. The DHA production potential from glycerol, ethanol and glucose is compared by combining fermentation experiments with the pathway-scale kinetic modeling and constraint-based stoichiometric modeling of C. cohnii metabolism. Glycerol has the slowest biomass growth rate among the tested substrates. This is partially compensated by the highest PUFAs fraction, where DHA is dominant. Mathematical modeling reveals that glycerol has the best experimentally observed carbon transformation rate into biomass, reaching the closest values to the theoretical upper limit. In addition to our observations, the published experimental evidence indicates that crude glycerol is readily consumed by C. cohnii, making glycerol an attractive substrate for DHA production.


Subject(s)
Dinoflagellida/metabolism , Docosahexaenoic Acids/metabolism , Models, Theoretical , Biomass , Ethanol/metabolism , Fermentation , Glucose/metabolism , Glycerol/metabolism
14.
Immunol Rev ; 281(1): 88-98, 2018 01.
Article in English | MEDLINE | ID: mdl-29247992

ABSTRACT

A shift in our understanding of macrophage biology has come about as a result of recent discoveries in the area of metabolic reprogramming of macrophages. The NLRP3 inflammasome drives the activation of caspase-1, leading to the production of IL-1ß, IL-18, and a type of cell death termed pyroptosis. The NLRP3 inflammasome has been shown to sense metabolites such as palmitate, uric acid, and cholesterol crystals and is inhibited by ketone bodies produced during metabolic flux. The NLRP3 inflammasome has also been shown to be regulated by mitochondrial reactive oxygen species and components of glycolysis, such as Hexokinase. Here, we review these findings and discuss their importance for inflammation and furthermore discuss potential therapeutic benefits of targeting NLRP3.


Subject(s)
Inflammasomes/metabolism , Macrophages/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Caspase 1/metabolism , Cellular Reprogramming , Cholesterol/metabolism , Hexokinase/metabolism , Humans , Ketone Bodies/metabolism , Palmitic Acid/metabolism , Pyroptosis , Reactive Oxygen Species/metabolism , Uric Acid/metabolism
15.
Angew Chem Int Ed Engl ; 61(51): e202212932, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36251920

ABSTRACT

Hydrogen (H2 ) is a geological source of reducing electrons that is thought to have powered the metabolism of the last universal common ancestor to all extant life, and that is still metabolized by various modern organisms. It has been suggested that H2 drove a geochemical analogue of some or all of the reverse Krebs cycle at the emergence of the metabolic network, catalyzed by metals, but this has yet to be demonstrated experimentally. Herein, we show that three consecutive steps of the reverse Krebs cycle, converting oxaloacetate into succinate, can be driven without enzymes and in one-pot by H2 as the reducing agent under mild conditions compatible with biological chemistry. Low catalytic amounts of nickel (10-20 mol %) or platinum group metals (0.1-1 mol %) or even small amounts of ground meteorites were found to promote the reductive chemistry at temperatures between 5 and 60 °C and over a wide pH range, including pH 7. These results lend additional support to the hypothesis that geologically produced hydrogen and metal catalysts could have initiated early metabolic networks.


Subject(s)
Hydrogen , Meteoroids , Hydrogen/chemistry , Citric Acid Cycle , Catalysis , Oxaloacetic Acid/chemistry , Metals
16.
J Biol Chem ; 295(39): 13410-13418, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32820045

ABSTRACT

An important context in which metabolism influences tumorigenesis is the genetic cancer syndrome hereditary leiomyomatosis and renal cell carcinoma (HLRCC), a disease in which mutation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) causes hyperaccumulation of fumarate. This electrophilic oncometabolite can alter gene activity at the level of transcription, via reversible inhibition of epigenetic dioxygenases, as well as posttranslationally, via covalent modification of cysteine residues. To better understand the potential for metabolites to influence posttranslational modifications important to tumorigenesis and cancer cell growth, here we report a chemoproteomic analysis of a kidney-derived HLRCC cell line. Using a general reactivity probe, we generated a data set of proteomic cysteine residues sensitive to the reduction in fumarate levels caused by genetic reintroduction of active FH into HLRCC cell lines. This revealed a broad up-regulation of cysteine reactivity upon FH rescue, which evidence suggests is caused by an approximately equal proportion of transcriptional and posttranslational modification-mediated regulation. Gene ontology analysis highlighted several new targets and pathways potentially modulated by FH mutation. Comparison of the new data set with prior studies highlights considerable heterogeneity in the adaptive response of cysteine-containing proteins in different models of HLRCC. This is consistent with emerging studies indicating the existence of cell- and tissue-specific cysteine-omes, further emphasizing the need for characterization of diverse models. Our analysis provides a resource for understanding the proteomic adaptation to fumarate accumulation and a foundation for future efforts to exploit this knowledge for cancer therapy.


Subject(s)
Cysteine/metabolism , Fumarate Hydratase/metabolism , Fumarates/metabolism , Kidney Neoplasms/metabolism , Leiomyomatosis/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Skin Neoplasms/metabolism , Uterine Neoplasms/metabolism , Cell Line, Tumor , Cysteine/genetics , Fumarate Hydratase/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
17.
J Biol Chem ; 295(24): 8155-8163, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32152229

ABSTRACT

Metabolite transport across cellular membranes is required for bioenergetic processes and metabolic signaling. The solute carrier family 13 (slc13) transporters mediate transport of the metabolites succinate and citrate and hence are of paramount physiological importance. Nevertheless, the mechanisms of slc13 transport and regulation are poorly understood. Here, a dynamic structural slc13 model suggested that an interfacial helix, H4c, which is common to all slc13s, stabilizes the stationary scaffold domain by anchoring it to the membrane, thereby facilitating movement of the SLC13 catalytic domain. Moreover, we found that intracellular determinants interact with the H4c anchor domain to modulate transport. This dual function is achieved by basic residues that alternately face either the membrane phospholipids or the intracellular milieu. This mechanism was supported by several experimental findings obtained using biochemical methods, electrophysiological measurements in Xenopus oocytes, and fluorescent microscopy of mammalian cells. First, a positively charged and highly conserved H4c residue, Arg108, was indispensable and crucial for metabolite transport. Furthermore, neutralization of other H4c basic residues inhibited slc13 transport function, thus mimicking the inhibitory effect of the slc13 inhibitor, slc26a6. Our findings suggest that the positive charge distribution across H4c domain controls slc13 transporter function and is utilized by slc13-interacting proteins in the regulation of metabolite transport.


Subject(s)
Metabolome , Symporters/chemistry , Symporters/metabolism , Amino Acid Sequence , Animals , Biological Transport , Citrates/metabolism , Conserved Sequence , HEK293 Cells , Humans , Lipid Bilayers/metabolism , Models, Biological , Molecular Dynamics Simulation , Mutant Proteins , Protein Domains , Structure-Activity Relationship , Xenopus laevis
18.
J Biol Chem ; 295(2): 375-389, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31776190

ABSTRACT

The SIN3 histone-modifying complex regulates the expression of multiple methionine catabolic genes, including SAM synthetase (Sam-S), as well as SAM levels. To further dissect the relationship between methionine catabolism and epigenetic regulation by SIN3, we sought to identify genes and metabolic pathways controlled by SIN3 and SAM synthetase (SAM-S) in Drosophila melanogaster Using several approaches, including RNAi-mediated gene silencing, RNA-Seq- and quantitative RT-PCR-based transcriptomics, and ultra-high-performance LC-MS/MS- and GC/MS-based metabolomics, we found that, as a global transcriptional regulator, SIN3 impacted a wide range of genes and pathways. In contrast, SAM-S affected only a narrow range of genes and pathways. The expression and levels of additional genes and metabolites, however, were altered in Sin3A+Sam-S dual knockdown cells. This analysis revealed that SIN3 and SAM-S regulate overlapping pathways, many of which involve one-carbon and central carbon metabolisms. In some cases, the factors acted independently; in some others, redundantly; and for a third set, in opposition. Together, these results, obtained from experiments with the chromatin regulator SIN3 and the metabolic enzyme SAM-S, uncover a complex relationship between metabolism and epigenetic regulation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Methionine Adenosyltransferase/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epigenesis, Genetic , Gene Regulatory Networks , Metabolome , Methionine Adenosyltransferase/genetics , RNA Interference , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Transcriptional Activation
19.
J Biol Chem ; 295(52): 18524-18538, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33087444

ABSTRACT

The divalent anion sodium symporter (DASS) family (SLC13) plays critical roles in metabolic homeostasis, influencing many processes, including fatty acid synthesis, insulin resistance, and adiposity. DASS transporters catalyze the Na+-driven concentrative uptake of Krebs cycle intermediates and sulfate into cells; disrupting their function can protect against age-related metabolic diseases and can extend lifespan. An inward-facing crystal structure and an outward-facing model of a bacterial DASS family member, VcINDY from Vibrio cholerae, predict an elevator-like transport mechanism involving a large rigid body movement of the substrate-binding site. How substrate binding influences the conformational state of VcINDY is currently unknown. Here, we probe the interaction between substrate binding and protein conformation by monitoring substrate-induced solvent accessibility changes of broadly distributed positions in VcINDY using a site-specific alkylation strategy. Our findings reveal that accessibility to all positions tested is modulated by the presence of substrates, with the majority becoming less accessible in the presence of saturating concentrations of both Na+ and succinate. We also observe separable effects of Na+ and succinate binding at several positions suggesting distinct effects of the two substrates. Furthermore, accessibility changes to a solely succinate-sensitive position suggests that substrate binding is a low-affinity, ordered process. Mapping these accessibility changes onto the structures of VcINDY suggests that Na+ binding drives the transporter into an as-yet-unidentified conformational state, involving rearrangement of the substrate-binding site-associated re-entrant hairpin loops. These findings provide insight into the mechanism of VcINDY, which is currently the only structurally characterized representative of the entire DASS family.


Subject(s)
Dicarboxylic Acid Transporters/chemistry , Dicarboxylic Acid Transporters/metabolism , Protein Conformation , Sodium/metabolism , Solvents/chemistry , Vibrio cholerae/metabolism , Binding Sites , Biological Transport , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Vibrio cholerae/growth & development
20.
J Biol Chem ; 295(27): 8988-8998, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32376686

ABSTRACT

DEAD-box helicase 5 (DDX5) is a founding member of the DEAD-box RNA helicase family, a group of enzymes that regulate ribonucleoprotein formation and function in every aspect of RNA metabolism, ranging from synthesis to decay. Our laboratory previously found that DDX5 is involved in energy homeostasis, a process that is altered in many cancers. Small cell lung cancer (SCLC) is an understudied cancer type for which effective treatments are currently unavailable. Using an array of methods, including short hairpin RNA-mediated gene silencing, RNA and ChIP sequencing analyses, and metabolite profiling, we show here that DDX5 is overexpressed in SCLC cell lines and that its down-regulation results in various metabolic and cellular alterations. Depletion of DDX5 resulted in reduced growth and mitochondrial dysfunction in the chemoresistant SCLC cell line H69AR. The latter was evidenced by down-regulation of genes involved in oxidative phosphorylation and by impaired oxygen consumption. Interestingly, DDX5 depletion specifically reduced intracellular succinate, a TCA cycle intermediate that serves as a direct electron donor to mitochondrial complex II. We propose that the oncogenic role of DDX5, at least in part, manifests as up-regulation of respiration supporting the energy demands of cancer cells.


Subject(s)
DEAD-box RNA Helicases/metabolism , Mitochondria/metabolism , Small Cell Lung Carcinoma/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , DEAD-box RNA Helicases/physiology , Humans , Mitochondria/physiology , Oligonucleotide Array Sequence Analysis/methods , RNA Helicases/metabolism , RNA, Small Interfering/metabolism , Ribonucleoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL