Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 319
Filter
1.
Sheng Wu Gong Cheng Xue Bao ; 40(9): 3025-3038, 2024 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-39319722

ABSTRACT

Guanidinoacetic acid, as an energetic substance, has a wide range of applications in the food, pharmaceutical, and feed industries. However, the biosynthesis of guanidinoacetic acid has not been applied in industrial production. In this study, we designed the synthetic route of guanidinoacetic acid in a food-grade strain of Bacillus subtilis. By regulating the expression of key enzymes, lifting feedback inhibition, and increasing membrane permeability, we achieved the efficient synthesis of guanidinoacetic acid by whole-cell catalysis. Firstly, the optimal L-arginine:glycine amidinotransferase was screened based on the phylogenetic tree, and the expression of the key enzyme was enhanced by a strategy combining strong promoter and genome integration. Secondly, the ornithine cycle for L-arginine synthesis in Corynebacterium glutamicum was introduced to alleviate the feedback inhibition of the enzyme by the byproduct L-ornithine, and the L-arginine degradation pathway was knocked down to enhance substrate regeneration. Thirdly, the expression of N-acetylmuramoyl-L-alanine amidase (LytC) was up-regulated to increase the cell membrane permeability. Finally, after optimization of whole-cell production conditions, strain Bs-13 achieved guanidinoacetic acid production at a titer of 13.1 g/L after 24 h, with a proudction rate of 0.54 g/(L·h) and a glycine conversion rate of 92.7%. The above strategy improved the production of guanidinoacetic acid and provided a reference for the biosynthesis of guanidinoacetic acid.


Subject(s)
Arginine , Bacillus subtilis , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Arginine/biosynthesis , Arginine/metabolism , Glycine/analogs & derivatives , Glycine/metabolism , Glycine/biosynthesis , Amidinotransferases/genetics , Amidinotransferases/metabolism , Corynebacterium glutamicum/metabolism , Corynebacterium glutamicum/genetics , N-Acetylmuramoyl-L-alanine Amidase/genetics , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Metabolic Engineering , Ornithine/biosynthesis , Ornithine/metabolism
2.
Science ; 385(6705): 174-178, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38991083

ABSTRACT

One of the hallmarks of living organisms is their capacity for self-organization and regeneration, which requires a tight integration of metabolic and genetic networks. We sought to construct a linked metabolic and genetic network in vitro that shows such lifelike behavior outside of a cellular context and generates its own building blocks from nonliving matter. We integrated the metabolism of the crotonyl-CoA/ethyl-malonyl-CoA/hydroxybutyryl-CoA cycle with cell-free protein synthesis using recombinant elements. Our network produces the amino acid glycine from CO2 and incorporates it into target proteins following DNA-encoded instructions. By orchestrating ~50 enzymes we established a basic cell-free operating system in which genetically encoded inputs into a metabolic network are programmed to activate feedback loops allowing for self-integration and (partial) self-regeneration of the complete system.


Subject(s)
Carbon Dioxide , Cell-Free System , Glycine , Metabolic Networks and Pathways , Protein Biosynthesis , Acyl Coenzyme A/metabolism , Carbon Dioxide/metabolism , Feedback, Physiological , Gene Regulatory Networks , Glycine/biosynthesis , Glycine/genetics
3.
Eur Heart J ; 43(36): 3477-3489, 2022 09 21.
Article in English | MEDLINE | ID: mdl-35728000

ABSTRACT

AIMS: Genetic dilated cardiomyopathy (DCM) is a leading cause of heart failure. Despite significant progress in understanding the genetic aetiologies of DCM, the molecular mechanisms underlying the pathogenesis of familial DCM remain unknown, translating to a lack of disease-specific therapies. The discovery of novel targets for the treatment of DCM was sought using phenotypic sceening assays in induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) that recapitulate the disease phenotypes in vitro. METHODS AND RESULTS: Using patient-specific iPSCs carrying a pathogenic TNNT2 gene mutation (p.R183W) and CRISPR-based genome editing, a faithful DCM model in vitro was developed. An unbiased phenotypic screening in TNNT2 mutant iPSC-derived cardiomyocytes (iPSC-CMs) with small molecule kinase inhibitors (SMKIs) was performed to identify novel therapeutic targets. Two SMKIs, Gö 6976 and SB 203580, were discovered whose combinatorial treatment rescued contractile dysfunction in DCM iPSC-CMs carrying gene mutations of various ontologies (TNNT2, TTN, LMNA, PLN, TPM1, LAMA2). The combinatorial SMKI treatment upregulated the expression of genes that encode serine, glycine, and one-carbon metabolism enzymes and significantly increased the intracellular levels of glucose-derived serine and glycine in DCM iPSC-CMs. Furthermore, the treatment rescued the mitochondrial respiration defects and increased the levels of the tricarboxylic acid cycle metabolites and ATP in DCM iPSC-CMs. Finally, the rescue of the DCM phenotypes was mediated by the activating transcription factor 4 (ATF4) and its downstream effector genes, phosphoglycerate dehydrogenase (PHGDH), which encodes a critical enzyme of the serine biosynthesis pathway, and Tribbles 3 (TRIB3), a pseudokinase with pleiotropic cellular functions. CONCLUSIONS: A phenotypic screening platform using DCM iPSC-CMs was established for therapeutic target discovery. A combination of SMKIs ameliorated contractile and metabolic dysfunction in DCM iPSC-CMs mediated via the ATF4-dependent serine biosynthesis pathway. Together, these findings suggest that modulation of serine biosynthesis signalling may represent a novel genotype-agnostic therapeutic strategy for genetic DCM.


Subject(s)
Cardiomyopathy, Dilated , Molecular Targeted Therapy , Myocytes, Cardiac , Protein Kinase Inhibitors , Serine , Troponin T , Activating Transcription Factor 4/metabolism , Adenosine Triphosphate/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Carbazoles/pharmacology , Carbazoles/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/genetics , Drug Evaluation, Preclinical/methods , Glucose/metabolism , Glycine/biosynthesis , Glycine/genetics , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Induced Pluripotent Stem Cells/physiology , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphoglycerate Dehydrogenase/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Serine/antagonists & inhibitors , Serine/biosynthesis , Serine/genetics , Troponin T/genetics , Troponin T/metabolism
4.
J Biol Chem ; 298(3): 101657, 2022 03.
Article in English | MEDLINE | ID: mdl-35131263

ABSTRACT

A wide range of bacteria possess virulence factors such as aminoacyl-tRNA transferases (ATTs) that are capable of rerouting aminoacyl-transfer RNAs away from protein synthesis to conjugate amino acids onto glycerolipids. We recently showed that, although these pathways were thought to be restricted to bacteria, higher fungi also possess ergosteryl-3ß-O-L-aspartate synthases (ErdSs), which transfer the L-Asp moiety of aspartyl-tRNAAsp onto the 3ß-OH group of ergosterol (Erg), yielding ergosteryl-3ß-O-L-aspartate (Erg-Asp). Here, we report the discovery, in fungi, of a second type of fungal sterol-specific ATTs, namely, ergosteryl-3ß-O-glycine (Erg-Gly) synthase (ErgS). ErgS consists of a freestanding DUF2156 domain encoded by a gene distinct from and paralogous to that of ErdS. We show that the enzyme only uses Gly-tRNAGly produced by an independent glycyl-tRNA synthetase (GlyRS) to transfer glycine onto the 3ß-OH of Erg, producing Erg-Gly. Phylogenomics analysis also show that the Erg-Gly synthesis pathway exists only in Ascomycota, including species of biotechnological interest, and more importantly, in human pathogens, such as Aspergillus fumigatus. The discovery of a second type of Erg-aa not only expands the repertoire of this particular class of fungal lipids but suggests that Erg-aa synthases might constitute a genuine subfamily of lipid-modifying ATTs.


Subject(s)
Ascomycota , Ergosterol , Glycine , Amino Acids , Ascomycota/genetics , Ascomycota/metabolism , Aspartic Acid , Glycine/biosynthesis , Glycine/genetics , Glycine/metabolism , Humans , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism
5.
Int J Oncol ; 58(2): 158-170, 2021 02.
Article in English | MEDLINE | ID: mdl-33491748

ABSTRACT

Serine/glycine biosynthesis and one­carbon metabolism are crucial in sustaining cancer cell survival and rapid proliferation, and of high clinical relevance. Excessive activation of serine/glycine biosynthesis drives tumorigenesis and provides a single carbon unit for one­carbon metabolism. One­carbon metabolism, which is a complex cyclic metabolic network based on the chemical reaction of folate compounds, provides the necessary proteins, nucleic acids, lipids and other biological macromolecules to support tumor growth. Moreover, one­carbon metabolism also maintains the redox homeostasis of the tumor microenvironment and provides substrates for the methylation reaction. The present study reviews the role of key enzymes with tumor­promoting functions and important intermediates that are physiologically relevant to tumorigenesis in serine/glycine/one­carbon metabolism pathways. The related regulatory mechanisms of action of the key enzymes and important intermediates in tumors are also discussed. It is hoped that investigations into these pathways will provide new translational opportunities for human cancer drug development, dietary interventions, and biomarker identification.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbon/metabolism , Glycine/biosynthesis , Neoplasms/pathology , Serine/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Disease Models, Animal , Humans , Metabolic Networks and Pathways/drug effects , Methylation/drug effects , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidation-Reduction/drug effects , Tumor Microenvironment/drug effects , Warburg Effect, Oncologic/drug effects
6.
Mol Cancer Ther ; 20(1): 50-63, 2021 01.
Article in English | MEDLINE | ID: mdl-33203732

ABSTRACT

Metabolic rewiring is a hallmark of cancer that supports tumor growth, survival, and chemotherapy resistance. Although normal cells often rely on extracellular serine and glycine supply, a significant subset of cancers becomes addicted to intracellular serine/glycine synthesis, offering an attractive drug target. Previously developed inhibitors of serine/glycine synthesis enzymes did not reach clinical trials due to unfavorable pharmacokinetic profiles, implying that further efforts to identify clinically applicable drugs targeting this pathway are required. In this study, we aimed to develop therapies that can rapidly enter the clinical practice by focusing on drug repurposing, as their safety and cost-effectiveness have been optimized before. Using a yeast model system, we repurposed two compounds, sertraline and thimerosal, for their selective toxicity against serine/glycine synthesis-addicted breast cancer and T-cell acute lymphoblastic leukemia cell lines. Isotope tracer metabolomics, computational docking, enzymatic assays, and drug-target interaction studies revealed that sertraline and thimerosal inhibit serine/glycine synthesis enzymes serine hydroxymethyltransferase and phosphoglycerate dehydrogenase, respectively. In addition, we demonstrated that sertraline's antiproliferative activity was further aggravated by mitochondrial inhibitors, such as the antimalarial artemether, by causing G1-S cell-cycle arrest. Most notably, this combination also resulted in serine-selective antitumor activity in breast cancer mouse xenografts. Collectively, this study provides molecular insights into the repurposed mode-of-action of the antidepressant sertraline and allows to delineate a hitherto unidentified group of cancers being particularly sensitive to treatment with sertraline. Furthermore, we highlight the simultaneous inhibition of serine/glycine synthesis and mitochondrial metabolism as a novel treatment strategy for serine/glycine synthesis-addicted cancers.


Subject(s)
Antidepressive Agents/pharmacology , Breast Neoplasms/pathology , Drug Repositioning , Glycine Hydroxymethyltransferase/antagonists & inhibitors , Glycine/biosynthesis , Serine/blood , Sertraline/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Glycine Hydroxymethyltransferase/metabolism , Humans , Mice, Inbred NOD , Mice, SCID , Molecular Docking Simulation , Phosphoglycerate Dehydrogenase/metabolism , Thimerosal/pharmacology
7.
FEMS Microbiol Lett ; 367(24)2020 01 15.
Article in English | MEDLINE | ID: mdl-33296465

ABSTRACT

The biochemical routes for assimilation of one-carbon compounds in bacteria require many clarifications. In this study, the role of malyl-CoA lyase in the metabolism of the aerobic type I methanotroph Methylotuvimicrobium alcaliphilum 20Z has been investigated by gene inactivation and biochemical studies. The functionality of the enzyme has been confirmed by heterologous expression in Escherichia coli. The mutant strain lacking Mcl activity demonstrated the phenotype of glycine auxotrophy. The genes encoding malyl-CoA lyase are present in the genomes of all methanotrophs, except for representatives of the phylum Verrucomicrobium. We suppose that malyl-CoA lyase is the enzyme that provides glyoxylate and glycine synthesis in the type I methanotrophs supporting carbon assimilation via the serine cycle in addition to the major ribulose monophosphate cycle.


Subject(s)
Bacterial Proteins/metabolism , Glycine/biosynthesis , Glyoxylates/metabolism , Methylococcaceae/enzymology , Oxo-Acid-Lyases/metabolism , Escherichia coli/genetics , Methylococcaceae/genetics
8.
Nature ; 586(7831): 790-795, 2020 10.
Article in English | MEDLINE | ID: mdl-32788725

ABSTRACT

Serine, glycine and other nonessential amino acids are critical for tumour progression, and strategies to limit their availability are emerging as potential therapies for cancer1-3. However, the molecular mechanisms driving this response remain unclear and the effects on lipid metabolism are relatively unexplored. Serine palmitoyltransferase (SPT) catalyses the de novo biosynthesis of sphingolipids but also produces noncanonical 1-deoxysphingolipids when using alanine as a substrate4,5. Deoxysphingolipids accumulate in the context of mutations in SPTLC1 or SPTLC26,7-or in conditions of low serine availability8,9-to drive neuropathy, and deoxysphinganine has previously been investigated as an anti-cancer agent10. Here we exploit amino acid metabolism and the promiscuity of SPT to modulate the endogenous synthesis of toxic deoxysphingolipids and slow tumour progression. Anchorage-independent growth reprogrammes a metabolic network involving serine, alanine and pyruvate that drives the endogenous synthesis and accumulation of deoxysphingolipids. Targeting the mitochondrial pyruvate carrier promotes alanine oxidation to mitigate deoxysphingolipid synthesis and improve spheroid growth, similar to phenotypes observed with the direct inhibition of SPT or ceramide synthesis. Restriction of dietary serine and glycine potently induces the accumulation of deoxysphingolipids while decreasing tumour growth in xenograft models in mice. Pharmacological inhibition of SPT rescues xenograft growth in mice fed diets restricted in serine and glycine, and the reduction of circulating serine by inhibition of phosphoglycerate dehydrogenase (PHGDH) leads to the accumulation of deoxysphingolipids and mitigates tumour growth. The promiscuity of SPT therefore links serine and mitochondrial alanine metabolism to membrane lipid diversity, which further sensitizes tumours to metabolic stress.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Serine/deficiency , Sphingolipids/chemistry , Sphingolipids/metabolism , Alanine/biosynthesis , Alanine/metabolism , Alanine/pharmacology , Animals , Cell Adhesion/drug effects , Cell Division/drug effects , Diet , Female , Glycine/biosynthesis , Glycine/deficiency , Glycine/metabolism , Glycine/pharmacology , HCT116 Cells , Humans , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Mice , Mitochondria/metabolism , Neoplasms/drug therapy , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Phosphoglycerate Dehydrogenase/metabolism , Pyruvic Acid/metabolism , Serine/blood , Serine/pharmacology , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/metabolism , Spheroids, Cellular/pathology , Sphingolipids/biosynthesis , Stress, Physiological/drug effects , Xenograft Model Antitumor Assays
9.
Enzyme Microb Technol ; 136: 109513, 2020 May.
Article in English | MEDLINE | ID: mdl-32331718

ABSTRACT

L-Phenylglycine (L-PHG) is a member of unnatural amino acids, and becoming more and more important as intermediate for pharmaceuticals, food additives and agrochemicals. However, the existing synthetic methods for L-PHG mainly rely on toxic cyanide chemistry and multistep processes. To provide green, safe and high enantioselective alternatives, we envisaged cascade biocatalysis for the one-pot synthesis of L-PHG from racemic mandelic acid. A engineered E. coli strain was established to co-express mandelate racemase, D-mandelate dehydrogenase and L-leucine dehydrogenase and catalyze a 3-step reaction in one pot, enantioselectively transforming racemic mandelic acid to give L-PHG (e.e. >99 %). After the conditions for biosynthesis of L-PHG optimized by response surface methodology, the yield and space-time yield of L-PHG can reach 87.89 % and 79.70 g·L-1·d-1, which was obviously improved. The high-yielding and enantioselective synthetic methods use cheap and green reagents, and E. coli whole-cell catalysts, thus providing green and useful alternative methods for manufacturing L-PHG.


Subject(s)
Glycine/analogs & derivatives , Industrial Microbiology/methods , Mandelic Acids/metabolism , Bacterial Proteins/metabolism , Biocatalysis , Escherichia coli/genetics , Escherichia coli/metabolism , Glycine/biosynthesis , Kinetics , Plasmids/genetics , Stereoisomerism
10.
Appl Microbiol Biotechnol ; 104(8): 3433-3444, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32078019

ABSTRACT

L-phenylglycine (L-Phg) is a rare non-proteinogenic amino acid, which only occurs in some natural compounds, such as the streptogramin antibiotics pristinamycin I and virginiamycin S or the bicyclic peptide antibiotic dityromycin. Industrially, more interesting than L-Phg is the enantiomeric D-Phg as it plays an important role in the fine chemical industry, where it is used as a precursor for the production of semisynthetic ß-lactam antibiotics. Based on the natural L-Phg operon from Streptomyces pristinaespiralis and the stereo-inverting aminotransferase gene hpgAT from Pseudomonas putida, an artificial D-Phg operon was constructed. The natural L-Phg operon, as well as the artificial D-Phg operon, was heterologously expressed in different actinomycetal host strains, which led to the successful production of Phg. By rational genetic engineering of the optimal producer strains S. pristinaespiralis and Streptomyces lividans, Phg production could be improved significantly. Here, we report on the development of a synthetic biology-derived D-Phg pathway and the optimization of fermentative Phg production in actinomycetes by genetic engineering approaches. Our data illustrate a promising alternative for the production of Phgs.


Subject(s)
Fermentation , Genetic Engineering/methods , Glycine/analogs & derivatives , Operon , Streptomyces lividans/genetics , Streptomyces/genetics , Anti-Bacterial Agents/biosynthesis , Genes, Bacterial , Glycine/biosynthesis , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Stereoisomerism , Synthetic Biology/methods
11.
Appl Microbiol Biotechnol ; 103(21-22): 8839-8851, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31642949

ABSTRACT

D-p-hydroxyphenylglycine (D-HPG) functions as an intermediate and has important value in antibiotic industries. The high pollution and costs from chemical processes make biotechnological route for D-HPG highly desirable. Here, a whole-cell transformation process by D-hydantoinase(Hase) and D-carbamoylase(Case) was developed to produce D-HPG from DL-hydroxyphenylhydantoin(DL-HPH) in Escherichia coli. The artificially designed ribosome binding site with strong intensity significantly facilitated the protein expression of limiting step enzyme Case. Next, the cell wall permeability was improved by disturbing the peptidoglycan structure by overproduction of D,D-carboxypeptidases without obviously affecting cell growth, to increase the bioavailability of low soluble hydantoin substrate. By fine-tuning regulation of expression level of D,D-carboxypeptidase DacB, the final production yield of D-HPG increased to 100% with 140 mM DL-HPH substrate under the optimized transformation conditions. This is the first example to enhance bio-productivity of chemicals by cell wall engineering and creates a new vision on biotransformation of sparingly soluble substrates. Additionally, the newly demonstrated 'hydroxyl occupancy' phenomenon when Case reacts with hydroxyl substrates provides a referential information for the enzyme engineering in future.


Subject(s)
Bioreactors/microbiology , Cell Engineering/methods , Cell Wall/genetics , Escherichia coli/metabolism , Glycine/analogs & derivatives , Protein Biosynthesis/genetics , Amidohydrolases/genetics , Amidohydrolases/metabolism , Carboxypeptidases/metabolism , Cell Wall/metabolism , Escherichia coli/genetics , Genetic Engineering , Glycine/biosynthesis , Permeability , Protein Biosynthesis/physiology
12.
J Biol Chem ; 294(36): 13464-13477, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31337706

ABSTRACT

Nucleotide synthesis is essential to proliferating cells, but the preferred precursors for de novo biosynthesis are not defined in human cancer tissues. We have employed multiplexed stable isotope-resolved metabolomics to track the metabolism of [13C6]glucose, D2-glycine, [13C2]glycine, and D3-serine into purine nucleotides in freshly resected cancerous and matched noncancerous lung tissues from nonsmall cell lung cancer (NSCLC) patients, and we compared the metabolism with established NSCLC PC9 and A549 cell lines in vitro Surprisingly, [13C6]glucose was the best carbon source for purine synthesis in human NSCLC tissues, in contrast to the noncancerous lung tissues from the same patient, which showed lower mitotic indices and MYC expression. We also observed that D3-Ser was preferentially incorporated into purine rings over D2-glycine in both tissues and cell lines. MYC suppression attenuated [13C6]glucose, D3-serine, and [13C2]glycine incorporation into purines and reduced proliferation in PC9 but not in A549 cells. Using detailed kinetic modeling, we showed that the preferred use of glucose as a carbon source for purine ring synthesis in NSCLC tissues involves cytoplasmic activation/compartmentation of the glucose-to-serine pathway and enhanced reversed one-carbon fluxes that attenuate exogenous serine incorporation into purines. Our findings also indicate that the substrate for de novo nucleotide synthesis differs profoundly between cancer cell lines and fresh human lung cancer tissues; the latter preferred glucose to exogenous serine or glycine but not the former. This distinction in substrate utilization in purine synthesis in human cancer tissues should be considered when targeting one-carbon metabolism for cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Glycine/biosynthesis , Lung Neoplasms/metabolism , Purine Nucleotides/biosynthesis , Serine/biosynthesis , A549 Cells , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/pathology , Metabolomics
13.
Sci Signal ; 12(582)2019 05 21.
Article in English | MEDLINE | ID: mdl-31113850

ABSTRACT

The differentiation of fibroblasts into a transient population of highly activated, extracellular matrix (ECM)-producing myofibroblasts at sites of tissue injury is critical for normal tissue repair. Excessive myofibroblast accumulation and persistence, often as a result of a failure to undergo apoptosis when tissue repair is complete, lead to pathological fibrosis and are also features of the stromal response in cancer. Myofibroblast differentiation is accompanied by changes in cellular metabolism, including increased glycolysis, to meet the biosynthetic demands of enhanced ECM production. Here, we showed that transforming growth factor-ß1 (TGF-ß1), the key pro-fibrotic cytokine implicated in multiple fibrotic conditions, increased the production of activating transcription factor 4 (ATF4), the transcriptional master regulator of amino acid metabolism, to supply glucose-derived glycine to meet the amino acid requirements associated with enhanced collagen production in response to myofibroblast differentiation. We further delineated the signaling pathways involved and showed that TGF-ß1-induced ATF4 production depended on cooperation between canonical TGF-ß1 signaling through Smad3 and activation of mechanistic target of rapamycin complex 1 (mTORC1) and its downstream target eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1). ATF4, in turn, promoted the transcription of genes encoding enzymes of the de novo serine-glycine biosynthetic pathway and glucose transporter 1 (GLUT1). Our findings suggest that targeting the TGF-ß1-mTORC1-ATF4 axis may represent a novel therapeutic strategy for interfering with myofibroblast function in fibrosis and potentially in other conditions, including cancer.


Subject(s)
Activating Transcription Factor 4/metabolism , Collagen/biosynthesis , Glycine/biosynthesis , Mechanistic Target of Rapamycin Complex 1/metabolism , Serine/biosynthesis , Transforming Growth Factor beta1/pharmacology , Activating Transcription Factor 4/genetics , Biosynthetic Pathways/drug effects , Biosynthetic Pathways/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Signal Transduction/drug effects
15.
Appl Environ Microbiol ; 85(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30367006

ABSTRACT

Acylated amino acids function as important components of the cellular membrane in some bacteria. Biosynthesis is initiated by the N-acylation of the amino acid, and this is followed by subsequent O-acylation of the acylated molecule, resulting in the production of the mature diacylated amino acid lipid. In this study, we use both genetics and liquid chromatography-mass spectrometry (LC-MS) to characterize the biosynthesis and function of a diacylated glycine lipid (GL) species produced in Bacteroides thetaiotaomicron We, and others, have previously reported the identification of a gene, named glsB in this study, that encodes an N-acyltransferase activity responsible for the production of a monoacylated glycine called N-acyl-3-hydroxy-palmitoyl glycine (or commendamide). In all of the Bacteroidales genomes sequenced so far, the glsB gene is located immediately downstream from a gene, named glsA, that is also predicted to encode a protein with acyltransferase activity. We use LC-MS to show that the coexpression of glsB and glsA results in the production of GL in Escherichia coli We constructed a deletion mutant of the glsB gene in B. thetaiotaomicron, and we confirm that glsB is required for the production of GL in B. thetaiotaomicron Moreover, we show that glsB is important for the ability of B. thetaiotaomicron to adapt to stress and colonize the mammalian gut. Therefore, this report describes the genetic requirements for the biosynthesis of GL, a diacylated amino acid species that contributes to fitness in the human gut bacterium B. thetaiotaomicronIMPORTANCE The gut microbiome has an important role in both health and disease of the host. The mammalian gut microbiome is often dominated by bacteria from the Bacteroidales, an order that includes Bacteroides and Prevotella In this study, we have identified an acylated amino acid, called glycine lipid, produced by Bacteroides thetaiotaomicron, a beneficial bacterium originally isolated from the human gut. In addition to identifying the genes required for the production of glycine lipids, we show that glycine lipids have an important role during the adaptation of B. thetaiotaomicron to a number of environmental stresses, including exposure to either bile or air. We also show that glycine lipids are important for the normal colonization of the murine gut by B. thetaiotaomicron This work identifies glycine lipids as an important fitness determinant in B. thetaiotaomicron and therefore increases our understanding of the molecular mechanisms underpinning colonization of the mammalian gut by beneficial bacteria.


Subject(s)
Bacteroides thetaiotaomicron/growth & development , Genetic Fitness , Glycine/biosynthesis , Lipids/biosynthesis , Animals , Bacteroides thetaiotaomicron/genetics , Female , Germ-Free Life , Lipid Metabolism , Mice , Mice, Inbred C57BL
16.
Biol Chem ; 400(3): 289-297, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30291781

ABSTRACT

Site-specific bioconjugation strategies offer many possibilities for directed protein modifications. Among the various enzyme-based conjugation protocols, formylglycine-generating enzymes allow to posttranslationally introduce the amino acid Cα-formylglycine (FGly) into recombinant proteins, starting from cysteine or serine residues within distinct consensus motifs. The aldehyde-bearing FGly-residue displays orthogonal reactivity to all other natural amino acids and can, therefore, be used for site-specific labeling reactions on protein scaffolds. In this review, the state of research on catalytic mechanisms and consensus motifs of different formylglycine-generating enzymes, as well as labeling strategies and applications of FGly-based bioconjugations are presented.


Subject(s)
Glycine/analogs & derivatives , Sulfatases/metabolism , Glycine/biosynthesis , Glycine/chemistry , Glycine/metabolism , Humans , Models, Molecular , Molecular Structure , Sulfatases/chemistry
17.
Cell Rep ; 24(1): 27-37.e4, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29972788

ABSTRACT

Multiple sulfatase deficiency (MSD) is a fatal, inherited lysosomal storage disorder characterized by reduced activities of all sulfatases in patients. Sulfatases require a unique post-translational modification of an active-site cysteine to formylglycine that is catalyzed by the formylglycine-generating enzyme (FGE). FGE mutations that affect intracellular protein stability determine residual enzyme activity and disease severity in MSD patients. Here, we show that protein disulfide isomerase (PDI) plays a pivotal role in the recognition and quality control of MSD-causing FGE variants. Overexpression of PDI reduces the residual activity of unstable FGE variants, whereas inhibition of PDI function rescues the residual activity of sulfatases in MSD fibroblasts. Mass spectrometric analysis of a PDI+FGE variant covalent complex allowed determination of the molecular signature for FGE recognition by PDI. Our findings highlight the role of PDI as a disease modifier in MSD, which may also be relevant for other ER-associated protein folding pathologies.


Subject(s)
Endoplasmic Reticulum/metabolism , Glycine/analogs & derivatives , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Protein Folding , Amino Acid Sequence , Disulfides/metabolism , Enzyme Stability , Glycine/biosynthesis , Humans , Multiple Sulfatase Deficiency Disease/enzymology , Mutant Proteins/metabolism , Mutation/genetics , Peptides/chemistry
18.
Mol Carcinog ; 57(10): 1342-1357, 2018 10.
Article in English | MEDLINE | ID: mdl-29873416

ABSTRACT

Ewing sarcoma (EWS) is a soft tissue and bone tumor that occurs primarily in adolescents and young adults. In most cases of EWS, the chimeric transcription factor, EWS-FLI1 is the primary oncogenic driver. The epigenome of EWS cells reflects EWS-FLI1 binding and activation or repression of transcription. Here, we demonstrate that EWS-FLI1 positively regulates the expression of proteins required for serine-glycine biosynthesis and uptake of the alternative nutrient source glutamine. Specifically, we show that EWS-FLI1 activates expression of PHGDH, PSAT1, PSPH, and SHMT2. Using cell-based studies, we also establish that EWS cells are dependent on glutamine for cell survival and that EWS-FLI1 positively regulates expression of the glutamine transporter, SLC1A5 and two enzymes involved in the one-carbon cycle, MTHFD2 and MTHFD1L. Inhibition of serine-glycine biosynthesis in EWS cells impacts their redox state leading to an accumulation of reactive oxygen species, DNA damage, and apoptosis. Importantly, analysis of EWS primary tumor transcriptome data confirmed that the aforementioned genes we identified as regulated by EWS-FLI1 exhibit increased expression compared with normal tissues. Furthermore, retrospective analysis of an independent data set generated a significant stratification of the overall survival of EWS patients into low- and high-risk groups based on the expression of PHGDH, PSAT1, PSPH, SHMT2, SLC1A5, MTHFD2, and MTHFD1L. In summary, our study demonstrates that EWS-FLI1 reprograms the metabolism of EWS cells and that serine-glycine metabolism or glutamine uptake are potential targetable vulnerabilities in this tumor type.


Subject(s)
Glutamine/metabolism , Glycine/biosynthesis , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Serine/biosynthesis , Amino Acid Transport System ASC/genetics , Amino Acid Transport System ASC/metabolism , Aminohydrolases/genetics , Aminohydrolases/metabolism , Apoptosis/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival/genetics , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Oncogene Proteins, Fusion/genetics , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology
19.
J Gen Appl Microbiol ; 64(5): 203-211, 2018 Nov 09.
Article in English | MEDLINE | ID: mdl-29709901

ABSTRACT

The aquatic cyanobacterium Nostoc verrucosum forms macroscopic colonies in streams, and its appearance is superficially similar to that of the terrestrial cyanobacterium Nostoc commune. N. verrucosum is sensitive to desiccation, unlike N. commune, although these Nostoc cyanobacterial species share physiological features, including massive extracellular polysaccharide production and trehalose accumulation capability. In this study, water-soluble sunscreen pigments of mycosporine-like amino acids (MAAs) were characterized in N. verrucosum, and the mysABCD genes responsible for MAA biosynthesis in N. verrucosum and N. commune were compared. N. verrucosum produced porphyra-334 and shinorine, with porphyra-334 accounting for >90% of the total MAAs. Interestingly, porphyra-334 is an atypical cyanobacteial MAA, whereas shinorine is known as a common and dominant MAA in cyanobacteria. Porphyra-334 from N. verrucosum showed little or no radical scavenging activity in vitro, although the glycosylated derivatives of porphyra-334 from N. commune are potent radical scavengers. The presence of the mysABCD gene cluster in N. commune strain KU002 (genotype A) supported its porphyra-334 producing capability via the Nostoc-type mechanism, although the genotype A of N. commune mainly produces the arabinose-bound porphyra-334. The mysABC gene cluster was conserved in N. verrucosum, but the mysD gene was not included in the cluster. These results suggest that the mysABCD gene products are involved in the biosynthesis of porphyra-334 commonly in these Nostoc species, and that the genotype A of N. commune additionally acquired the glycosylation of porphyra-334.


Subject(s)
Cyclohexanones , Cyclohexylamines , Glycine/analogs & derivatives , Nostoc/chemistry , Cyclohexanones/metabolism , Cyclohexylamines/metabolism , Glycine/biosynthesis , Glycine/genetics , Glycine/metabolism , Glycosylation , Multigene Family/genetics , Nostoc/genetics , Sunscreening Agents/chemistry
20.
ACS Synth Biol ; 7(5): 1424-1435, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29609457

ABSTRACT

Metabolic engineering and synthetic biology usually require universal expression systems for stable and efficient gene expression in various organisms. In this study, a host-independent and stable T7 expression system had been developed by integrating T7 RNA polymerase and its cognate transcriptional units in single plasmid. The expression of T7 RNA polymerase was restricted below its lethal threshold using a T7 RNA polymerase antisense gene cassette, which allowed long periods of cultivation and protein production. In addition, by designing ribosome binding sites, we further tuned the expression capacity of this novel T7 system within a wide range. This host-independent expression system efficiently expressed genes in five different Gram-negative strains and one Gram-positive strain and was also shown to be applicable in a real industrial d- p-hydroxyphenylglycine production system.


Subject(s)
DNA-Directed RNA Polymerases/genetics , Glycine/analogs & derivatives , Metabolic Engineering/methods , Plasmids/genetics , Viral Proteins/genetics , Antisense Elements (Genetics) , Binding Sites , Corynebacterium glutamicum/genetics , DNA-Directed RNA Polymerases/metabolism , Gene Expression Regulation, Bacterial , Glycine/biosynthesis , Gram-Negative Bacteria/genetics , Microorganisms, Genetically-Modified , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosomes/metabolism , Sinorhizobium/genetics , Transcription, Genetic , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL