Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.061
Filter
1.
Methods Enzymol ; 704: 313-344, 2024.
Article in English | MEDLINE | ID: mdl-39300654

ABSTRACT

Aspartate/asparagine-ß-hydroxylase (AspH) is a transmembrane 2-oxoglutarate (2OG)-dependent oxygenase that catalyzes the post-translational hydroxylation of aspartate- and asparagine-residues in epidermal growth factor-like domains (EGFDs) of its substrate proteins. Upregulation of ASPH and translocation of AspH from the endoplasmic reticulum membrane to the surface membrane of cancer cells is associated with enhanced cell motility and worsened clinical prognosis. AspH is thus a potential therapeutic and diagnostic target for cancer. This chapter describes methods for the production and purification of soluble constructs of recombinant human AspH suitable for biochemical and crystallographic studies. The chapter also describes efficient methods for performing turnover and inhibition assays which monitor catalysis of isolated recombinant human AspH in vitro using solid phase extraction coupled to mass spectrometry (SPE-MS). The SPE-MS assays employ synthetic disulfide- or thioether-bridged macrocyclic oligopeptides as substrates; a macrocycle is an apparently essential requirement for productive AspH catalysis and mimics an EGFD disulfide isomer that is not typically observed in crystal and NMR structures. SPE-MS assays can be used to monitor catalysis of 2OG oxygenases other than AspH; the methods described herein are representative for 2OG oxygenase SPE-MS assays useful for performing kinetic and/or inhibition studies.


Subject(s)
Mixed Function Oxygenases , Recombinant Proteins , Humans , Recombinant Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/isolation & purification , Enzyme Assays/methods , Solid Phase Extraction/methods , Mass Spectrometry/methods , Catalysis , Kinetics , Asparagine/metabolism , Asparagine/chemistry , Hydroxylation , Substrate Specificity , Animals , Calcium-Binding Proteins , Membrane Proteins , Muscle Proteins
2.
Cell Rep Med ; 5(9): 101728, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39293390

ABSTRACT

Despite advances in treatment, the prognosis of patients with osteosarcoma remains unsatisfactory, and searching for potential targets is imperative. Here, we identify N4-acetylcytidine (ac4C) acetyltransferase 10 (NAT10) as a candidate therapeutic target in osteosarcoma through functional screening. NAT10 overexpression is correlated with a poor prognosis, and NAT10 knockout inhibits osteosarcoma progression. Mechanistically, NAT10 enhances mRNA stability of activating transcription factor 4 (ATF4) through ac4C modification. ATF4 induces the transcription of asparagine synthetase (ASNS), which catalyzes asparagine (Asn) biosynthesis, facilitating osteosarcoma progression. Utilizing virtual screening, we identify paliperidone and AG-401 as potential NAT10 inhibitors, and both inhibitors are found to bind to NAT10 proteins. Inhibiting NAT10 suppresses osteosarcoma progression in vivo. Combined treatment using paliperidone and AG-401 produces synergistic inhibition for osteosarcoma in patient-derived xenograft (PDX) models. Our findings demonstrate that NAT10 facilitates osteosarcoma progression through the ATF4/ASNS/Asn axis, and pharmacological inhibition of NAT10 may be a feasible therapeutic approach for osteosarcoma.


Subject(s)
Activating Transcription Factor 4 , Asparagine , Aspartate-Ammonia Ligase , Osteosarcoma , Humans , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Animals , Cell Line, Tumor , Aspartate-Ammonia Ligase/metabolism , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/antagonists & inhibitors , Mice , Asparagine/metabolism , Disease Progression , Xenograft Model Antitumor Assays , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects , Mice, Nude , Male , Female
3.
Acta Neuropathol Commun ; 12(1): 122, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164793

ABSTRACT

Gliomas, particularly glioblastomas (GBMs), pose significant challenges due to their aggressiveness and poor prognosis. Early detection through biomarkers is critical for improving outcomes. This study aimed to identify novel biomarkers for gliomas, particularly GBMs, using chiral amino acid profiling. We used chiral amino acid analysis to measure amino acid L- and D-isomer levels in resected tissues (tumor and non-tumor), blood, and urine from 33 patients with primary gliomas and 24 healthy volunteers. The levels of D-amino acid oxidase (DAO), a D-amino acid-degrading enzyme, were evaluated to investigate the D-amino acid metabolism in brain tissue. The GBM mouse model was created by transplanting GBM cells into the brain to confirm whether gliomas affect blood and urine chiral amino acid profiles. We also assessed whether D-amino acids produced by GBM cells are involved in cell proliferation. D-asparagine (D-Asn) levels were higher and DAO expression was lower in glioma than in non-glioma tissues. Blood and urinary D-Asn levels were lower in patients with GBM than in healthy volunteers (p < 0.001), increasing after GBM removal (p < 0.05). Urinary D-Asn levels differentiated between healthy volunteers and patients with GBM (area under the curve: 0.93, sensitivity: 0.88, specificity: 0.92). GBM mouse model validated the decrease of urinary D-Asn in GBM. GBM cells used D-Asn for cell proliferation. Gliomas induce alterations in chiral amino acid profiles, affecting blood and urine levels. Urinary D-Asn emerges as a promising diagnostic biomarker for gliomas, reflecting tumor presence and severity.


Subject(s)
Asparagine , Brain Neoplasms , D-Amino-Acid Oxidase , Glioblastoma , Humans , Glioblastoma/metabolism , Glioblastoma/urine , Glioblastoma/pathology , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/urine , Brain Neoplasms/pathology , Male , Middle Aged , Female , Asparagine/urine , Asparagine/metabolism , Adult , D-Amino-Acid Oxidase/metabolism , D-Amino-Acid Oxidase/genetics , Mice , Aged , Biomarkers, Tumor/urine , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Models, Animal , Cell Proliferation
4.
PLoS One ; 19(8): e0307448, 2024.
Article in English | MEDLINE | ID: mdl-39093903

ABSTRACT

Cancer is a leading concern and important cause of death worldwide. Cancer is a non-communicable illness defined as uncontrolled division of cells. It can develop into metastatic cancer when tumor cells migrate to other organs. In recent years evidence has emerged that the bioavailability of Asn play a crucial role in cancer metastasis. Asn is a non-essential amino acid formed from an ATP dependent catalyzed reaction by the enzyme asparagine synthetase (ASNS), where Asp and Gln are converted to Asn and Glu, respectively. The human ASNS enzyme consist of 561 amino acids, with a molecular weight of 64 KDa. ASNS governs the activation of transcriptional factors that regulate the process of metastasis. In this work the 3D model of ASNS in E. coli (AS-B) and the human ASNS docked with its different ligands have been used to study the 3D mechanism of the conversion of Asp and Gln to Asn and Glu, in human ASNS. The stability evaluation of the docked complexes was checked by molecular dynamic simulation through the bioinformatic tool Desmond. The binding residues and their interactions can be exploited for the development of inhibitors, as well as for finding new drug molecules against ASNS and prevention of metastatic cancer.


Subject(s)
Aspartate-Ammonia Ligase , Catalytic Domain , Molecular Dynamics Simulation , Humans , Aspartate-Ammonia Ligase/metabolism , Aspartate-Ammonia Ligase/chemistry , Aspartate-Ammonia Ligase/genetics , Molecular Docking Simulation , Substrate Specificity , Asparagine/metabolism , Asparagine/chemistry , Protein Binding , Escherichia coli/metabolism , Escherichia coli/genetics , Escherichia coli/enzymology , Computer Simulation , Ligands , Aspartic Acid/metabolism , Aspartic Acid/chemistry , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor
5.
FEBS Lett ; 598(18): 2269-2280, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39031546

ABSTRACT

SIRT5, one of the mammalian sirtuins, specifically recognizes succinyl-lysine residues on proteins and catalyzes the desuccinylation reaction. In this study, we characterized SIRT5 mutants with hydrophobic amino acid substitutions at Q140 and N141, in addition to the catalytic residue H158, known as an active site residue, by the Michaelis-Menten analysis and X-ray crystallography. Kinetic analysis showed that the catalytic efficiency (kcat/Km) of the Q140L and N141V mutants decreased to 0.02 times and 0.0038 times that of the wild-type SIRT5, respectively, with the activity of the N141V mutant becoming comparable to that of the H158M mutant. Our findings indicate that N141 contributes significantly to the desuccinylation reaction.


Subject(s)
Asparagine , Glutamine , NAD , Sirtuins , Asparagine/metabolism , Asparagine/chemistry , Asparagine/genetics , Sirtuins/metabolism , Sirtuins/genetics , Sirtuins/chemistry , Glutamine/metabolism , Glutamine/chemistry , Glutamine/genetics , Humans , Crystallography, X-Ray , NAD/metabolism , NAD/chemistry , Kinetics , Catalytic Domain , Binding Sites , Amino Acid Substitution , Models, Molecular , Mutation , Conserved Sequence , Animals
6.
Cancer Res ; 84(18): 3004-3022, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-38959335

ABSTRACT

TP53 tumor suppressor is frequently altered in lethal, castration-resistant prostate cancer (CRPC). However, to date there are no effective treatments that specifically target TP53 alterations. Using transcriptomic and metabolomic analyses, we have shown here that TP53-altered prostate cancer exhibits an increased dependency on asparagine (Asn) and overexpresses Asn synthetase (ASNS), the enzyme catalyzing the synthesis of Asn. Mechanistically, the loss or mutation of TP53 transcriptionally activated ASNS expression, directly and via mTORC1-mediated ATF4 induction, driving de novo Asn biosynthesis to support CRPC growth. TP53-altered CRPC cells were sensitive to Asn restriction by knockdown of ASNS or L-asparaginase treatment to deplete the intracellular and extracellular sources of Asn, respectively, and cell viability was rescued by Asn addition. Notably, pharmacological inhibition of intracellular Asn biosynthesis using a glutaminase inhibitor and depletion of extracellular Asn with L-asparaginase significantly reduced Asn production and effectively impaired CRPC growth. This study highlights the significance of ASNS-mediated metabolic adaptation as a synthetic vulnerability in CRPC with TP53 alterations, providing a rationale for targeting Asn production to treat these lethal prostate cancers. Significance: TP53-mutated castration-resistant prostate cancer is dependent on asparagine biosynthesis due to upregulation of ASNS and can be therapeutically targeted by approaches that deplete intracellular and extracellular asparagine.


Subject(s)
Asparagine , Prostatic Neoplasms, Castration-Resistant , Tumor Suppressor Protein p53 , Male , Humans , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Asparagine/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice , Animals , Cell Line, Tumor , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/metabolism , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Cell Proliferation , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor
7.
Pharmacol Res ; 206: 107292, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002867

ABSTRACT

Nutrient bioavailability in the tumor microenvironment plays a pivotal role in tumor proliferation and metastasis. Among these nutrients, glutamine is a key substance that promotes tumor growth and proliferation, and its downstream metabolite asparagine is also crucial in tumors. Studies have shown that when glutamine is exhausted, tumor cells can rely on asparagine to sustain their growth. Given the reliance of tumor cell proliferation on asparagine, restricting its bioavailability has emerged as promising strategy in cancer treatment. For instance, the use of asparaginase, an enzyme that depletes asparagine, has been one of the key chemotherapies for acute lymphoblastic leukemia (ALL). However, tumor cells can adapt to asparagine restriction, leading to reduced chemotherapy efficacy, and the mechanisms by which different genetically altered tumors are sensitized or adapted to asparagine restriction vary. We review the sources of asparagine and explore how limiting its bioavailability impacts the progression of specific genetically altered tumors. It is hoped that by targeting the signaling pathways involved in tumor adaptation to asparagine restriction and certain factors within these pathways, the issue of drug resistance can be addressed. Importantly, these strategies offer precise therapeutic approaches for genetically altered cancers.


Subject(s)
Asparagine , Neoplasms , Humans , Asparagine/metabolism , Animals , Neoplasms/drug therapy , Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Tumor Microenvironment/drug effects , Molecular Targeted Therapy
8.
Protein Sci ; 33(8): e5120, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39022918

ABSTRACT

Deamidation frequently is invoked as an important driver of crystallin aggregation and cataract formation. Here, we characterized the structural and biophysical consequences of cumulative Asn to Asp changes in γD-crystallin. Using NMR spectroscopy, we demonstrate that N- or C-terminal domain-confined or fully Asn to Asp changed γD-crystallin exhibits essentially the same 1H-15N HSQC spectrum as the wild-type protein, implying that the overall structure is retained. Only a very small thermodynamic destabilization for the overall Asn to Asp γD-crystallin variants was noted by chaotropic unfolding, and assessment of the colloidal stability, by measuring diffusion interaction parameters, yielded no substantive differences in association propensities. Furthermore, using molecular dynamics simulations, no significant changes in dynamics for proteins with Asn to Asp or iso-Asp changes were detected. Our combined results demonstrate that substitution of all Asn by Asp residues, reflecting an extreme case of deamidation, did not affect the structure and biophysical properties of γD-crystallin. This suggests that these changes alone cannot be the major determinant in driving cataract formation.


Subject(s)
Asparagine , Aspartic Acid , Molecular Dynamics Simulation , Protein Stability , gamma-Crystallins , gamma-Crystallins/chemistry , gamma-Crystallins/metabolism , gamma-Crystallins/genetics , Asparagine/chemistry , Asparagine/metabolism , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Thermodynamics , Cataract/metabolism , Cataract/genetics , Amino Acid Substitution
9.
Food Chem ; 458: 140285, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38970956

ABSTRACT

Sprouting can enhance the bioavailability and stimulate the production of health-promoting compounds. This research explored the potential health benefits of wheat sprouting, focusing on underexplored areas in existing literature such as alterations in phenylalanine ammonia-lyase (PAL) activity and glutathione levels during wheat sprouting. Furthermore, special attention was directed toward asparagine (Asn), the main precursor of acrylamide formation, as regulatory agencies are actively seeking to impose limitations on the presence of acrylamide in baked products. The results demonstrate elevated levels of PAL (4.5-fold at 48 h of sprouting), antioxidants, and total phenolics (1.32 mg gallic acid equivalent/g dry matter at 72 h of sprouting), coupled with a reduction in Asn (i.e. 11-fold at 48 h of sprouting) and glutathione concentrations, after wheat sprouting. These findings suggest that sprouting can unlock health-promoting properties in wheat. Optimizing the sprouting process to harness these benefits, however, may have implications for the techno-functionality of wheat flour in food processing.


Subject(s)
Flour , Phenylalanine Ammonia-Lyase , Triticum , Triticum/chemistry , Triticum/growth & development , Triticum/metabolism , Phenylalanine Ammonia-Lyase/metabolism , Phenylalanine Ammonia-Lyase/genetics , Flour/analysis , Germination , Antioxidants/metabolism , Antioxidants/chemistry , Plant Proteins/metabolism , Plant Proteins/genetics , Food Handling , Seeds/chemistry , Seeds/growth & development , Seeds/metabolism , Glutathione/metabolism , Asparagine/metabolism , Asparagine/chemistry , Acrylamide/metabolism , Acrylamide/chemistry , Phenols/metabolism , Phenols/chemistry
10.
PLoS One ; 19(7): e0306856, 2024.
Article in English | MEDLINE | ID: mdl-38991013

ABSTRACT

Site-specific modifications of aspartate residues spontaneously occur in crystallin, the major protein in the lens. One of the primary modification sites is Asp151 in αA-crystallin. Isomerization and racemization alter the crystallin backbone structure, reducing its stability by inducing abnormal crystallin-crystallin interactions and ultimately leading to the insolubilization of crystallin complexes. These changes are considered significant factors in the formation of senile cataracts. However, the mechanisms driving spontaneous isomerization and racemization have not been experimentally demonstrated. In this study, we generated αA-crystallins with different homo-oligomeric sizes and/or containing an asparagine residue at position 151, which is more prone to isomerization and racemization. We characterized their structure, hydrophobicity, chaperone-like function, and heat stability, and examined their propensity for isomerization and racemization. The results show that the two differently sized αA-crystallin variants possessed similar secondary structures but exhibited different chaperone-like functions depending on their oligomeric sizes. The rate of isomerization and racemization of Asp151, as assessed by the deamidation of Asn151, was also found to depend on the oligomeric sizes of αA-crystallin. The predominant isomerization product via deamidation of Asn151 in the different-sized αA-crystallin variants was L-ß-Asp in vitro, while various modifications occurred around Asp151 in vivo. The disparity between the findings of this in vitro study and in vivo studies suggests that the isomerization of Asp151 in vivo may be more complex than what occurs in vitro.


Subject(s)
Aspartic Acid , Protein Multimerization , alpha-Crystallin A Chain , Humans , alpha-Crystallin A Chain/chemistry , alpha-Crystallin A Chain/metabolism , alpha-Crystallin A Chain/genetics , Asparagine/chemistry , Asparagine/metabolism , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Hydrophobic and Hydrophilic Interactions , Isomerism , Protein Stability , Protein Structure, Secondary
11.
Med Oncol ; 41(7): 176, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879707

ABSTRACT

Asparagine is a non-essential amino acid crucial for protein biosynthesis and function, and therefore cell maintenance and growth. Furthermore, this amino acid has an important role in regulating several metabolic pathways, such as tricarboxylic acid cycle and the urea cycle. When compared to normal cells, tumor cells typically present a higher demand for asparagine, making it a compelling target for therapy. In this review article, we investigate different facets of asparagine bioavailability intricate role in malignant tumors raised from solid organs. We take a comprehensive look at asparagine synthetase expression and regulation in cancer, including the impact on tumor growth and metastasis. Moreover, we explore asparagine depletion through L-asparaginase as a potential therapeutic method for aggressive solid tumors, approaching different formulations of the enzyme and combinatory therapies. In summary, here we delve into studies about endogenous and exogenous asparagine availability in solid cancers, analyzing therapeutic implications and future challenges.


Subject(s)
Asparagine , Aspartate-Ammonia Ligase , Neoplasms , Humans , Asparagine/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/drug therapy , Aspartate-Ammonia Ligase/metabolism , Aspartate-Ammonia Ligase/genetics , Asparaginase/therapeutic use , Animals
12.
Res Microbiol ; 175(7): 104219, 2024.
Article in English | MEDLINE | ID: mdl-38945250

ABSTRACT

Despite being classified as microaerophilic microorganisms, most Campylobacter species can grow anaerobically, using formate or molecular hydrogen (H2) as electron donors, and various nitrogenous and sulfurous compounds as electron acceptors. Herein, we showed that both l-asparagine (l-Asn) and l-aspartic acid (l-Asp) bolster H2-driven anaerobic growth in several Campylobacter species, whereas the d-enantiomer form of both asparagine (d-Asn) and aspartic acid (d-Asp) only increased anaerobic growth in Campylobacter concisus strain 13826 and Campylobacter ureolyticus strain NCTC10941. A gene annotated as racD encoding for a putative d/l-Asp racemase was identified in the genome of both strains. Disruption of racD in Cc13826 resulted in the inability of the mutant strain to use either d-enantiomer during anaerobic growth. Hence, our results suggest that the racD gene is required for campylobacters to use either d-Asp or d-Asn. The use of d-Asp by various human opportunistic bacterial pathogens, including C. concisus, C. ureolyticus, and also possibly select strains of Campylobacter gracilis, Campylobacter rectus and Campylobacter showae, is significant, because d-Asp is an important signal molecule for both human nervous and neuroendocrine systems. To our knowledge, this is the first report of pathogens scavenging a d-amino acid essential for human health.


Subject(s)
Campylobacter , Campylobacter/genetics , Campylobacter/metabolism , Campylobacter/growth & development , Anaerobiosis , Humans , Amino Acid Isomerases/metabolism , Amino Acid Isomerases/genetics , Hydrogen/metabolism , D-Aspartic Acid/metabolism , Asparagine/metabolism
13.
J Mol Biol ; 436(16): 168666, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38880378

ABSTRACT

Heliorhodopsin (HeR) is a new rhodopsin family discovered in 2018 through functional metagenomic analysis. Similar to microbial rhodopsins, HeR has an all-trans retinal chromophore, and its photoisomerization to the 13-cis form triggers a relatively slow photocycle with sequential intermediate states (K, M, and O intermediates). The O intermediate has a relatively long lifetime and is a putative active state for transferring signals or regulating enzymatic reactions. Although the first discovered HeR, 48C12, was found in bacteria and the second HeR (TaHeR) was found in archaea, their key amino acid residues and molecular architectures have been recognized to be well conserved. Nevertheless, the rise and decay kinetics of the O intermediate are faster in 48C12 than in TaHeR. Here, using a new infrared spectroscopic technique with quantum cascade lasers, we clarified that the hydrogen bond between transmembrane helices (TM) 3 and 4 is essential for the altered O kinetics (Ser112 and Asn138 in 48C12). Interconverting mutants of 48C12 and TaHeR clearly revealed that the hydrogen bond is important for regulating the dynamics of the O intermediate. Overall, our study sheds light on the importance of the hydrogen bond between TM3 and TM4 in heliorhodopsins, similar to the DC gate in channelrhodopsins.


Subject(s)
Hydrogen Bonding , Kinetics , Rhodopsins, Microbial/chemistry , Rhodopsins, Microbial/metabolism , Rhodopsins, Microbial/genetics , Serine/chemistry , Serine/metabolism , Asparagine/chemistry , Asparagine/metabolism , Models, Molecular , Protein Conformation
14.
Amino Acids ; 56(1): 38, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844708

ABSTRACT

Biomarkers that accurately reflect renal function are essential in management of chronic kidney diseases (CKD). However, in children, age/physique and medication often alter established renal biomarkers. We studied whether amino acid enantiomers in body fluids correlate with renal function and whether they are influenced by physique or steroid medication during development. We conducted a prospective study of children 2 to 18 years old with and without CKD. We analyzed associations of serine/asparagine enantiomers in body fluids with major biochemical parameters as well as physique. To study consequences of kidney dysfunction and steroids on serine/asparagine enantiomers, we generated juvenile mice with uninephrectomy, ischemic reperfusion injury, or dexamethasone treatment. We obtained samples from 27 children, of which 12 had CKD due to congenital (n = 7) and perinatal (n = 5) causes. Plasma D-asparagine and the D/L-serine ratio had robust, positive linear associations with serum creatinine and cystatin C, and detected CKD with high sensitivity and specificity, uninfluenced by body size or biochemical parameters. In the animal study, kidney dysfunction increased plasma D-asparagine and the D/L-serine ratio, but dexamethasone treatment did not. Thus, plasma D-asparagine and the D/L-serine ratio can be useful markers for renal function in children.


Subject(s)
Asparagine , Biomarkers , Renal Insufficiency, Chronic , Serine , Child , Animals , Humans , Asparagine/blood , Asparagine/metabolism , Renal Insufficiency, Chronic/blood , Child, Preschool , Serine/blood , Mice , Male , Female , Adolescent , Biomarkers/blood , Prospective Studies , Dexamethasone , Stereoisomerism , Creatinine/blood , Kidney/metabolism
15.
Br J Haematol ; 205(1): 175-188, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38736325

ABSTRACT

B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) blasts strictly depend on the transport of extra-cellular asparagine (Asn), yielding a rationale for L-asparaginase (ASNase) therapy. However, the carriers used by ALL blasts for Asn transport have not been identified yet. Exploiting RS4;11 cells as BCP-ALL model, we have found that cell Asn is lowered by either silencing or inhibition of the transporters ASCT2 or SNAT5. The inhibitors V-9302 (for ASCT2) and GluγHA (for SNAT5) markedly lower cell proliferation and, when used together, suppress mTOR activity, induce autophagy and cause a severe nutritional stress, leading to a proliferative arrest and a massive cell death in both the ASNase-sensitive RS4;11 cells and the relatively ASNase-insensitive NALM-6 cells. The cytotoxic effect is not prevented by coculturing leukaemic cells with primary mesenchymal stromal cells. Leukaemic blasts of paediatric ALL patients express ASCT2 and SNAT5 at diagnosis and undergo marked cytotoxicity when exposed to the inhibitors. ASCT2 expression is positively correlated with the minimal residual disease at the end of the induction therapy. In conclusion, ASCT2 and SNAT5 are the carriers exploited by ALL cells to transport Asn, and ASCT2 expression is associated with a lower therapeutic response. ASCT2 may thus represent a novel therapeutic target in BCP-ALL.


Subject(s)
Amino Acid Transport System ASC , Asparagine , Cell Survival , Minor Histocompatibility Antigens , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Amino Acid Transport System ASC/metabolism , Amino Acid Transport System ASC/genetics , Asparagine/metabolism , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Cell Survival/drug effects , Amino Acid Transport System A/metabolism , Amino Acid Transport System A/genetics , Cell Line, Tumor , Asparaginase/pharmacology , Asparaginase/therapeutic use , Cell Proliferation/drug effects , Child
16.
Microbiol Immunol ; 68(5): 165-178, 2024 May.
Article in English | MEDLINE | ID: mdl-38444370

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is an ongoing global public health crisis. The causative agent, the SARS-CoV-2 virus, enters host cells via molecular interactions between the viral spike protein and the host cell ACE2 surface protein. The SARS-CoV-2 spike protein is extensively decorated with up to 66 N-linked glycans. Glycosylation of viral proteins is known to function in immune evasion strategies but may also function in the molecular events of viral entry into host cells. Here, we show that N-glycosylation at Asn331 and Asn343 of SARS-CoV-2 spike protein is required for it to bind to ACE2 and for the entry of pseudovirus harboring the SARS-CoV-2 spike protein into cells. Interestingly, high-content glycan binding screening data have shown that N-glycosylation of Asn331 and Asn343 of the RBD is important for binding to the specific glycan molecule G4GN (Galß-1,4 GlcNAc), which is critical for spike-RBD-ACE2 binding. Furthermore, IL-6 was identified through antibody array analysis of conditioned media of the corresponding pseudovirus assay. Mutation of N-glycosylation of Asn331 and Asn343 sites of the spike receptor-binding domain (RBD) significantly reduced the transcriptional upregulation of pro-inflammatory signaling molecule IL-6. In addition, IL-6 levels correlated with spike protein levels in COVID-19 patients' serum. These findings establish the importance of RBD glycosylation in SARS-CoV-2 pathogenesis, which can be exploited for the development of novel therapeutics for COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Interleukin-6 , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Internalization , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Humans , Glycosylation , Angiotensin-Converting Enzyme 2/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Interleukin-6/metabolism , COVID-19/virology , COVID-19/metabolism , HEK293 Cells , Asparagine/metabolism , Polysaccharides/metabolism
17.
Int J Biol Macromol ; 263(Pt 2): 130312, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38403216

ABSTRACT

L-Asparaginase is a key component in the treatment of leukemias and lymphomas. However, the glutamine affinity of this therapeutic enzyme is an off-target activity that causes several side effects. The modeling and molecular docking study of Yarrowia lipolytica L-asparaginase (YL-ASNase) to reduce its l-glutamine affinity and increase its stability was the aim of this study. Protein-ligand interactions of wild-type and different mutants of YL-ASNase against L-asparagine compared to l-glutamine were assessed using AutoDock Vina tools because the crystal structure of YL-ASNase does not exist in the protein data banks. The results showed that three mutants, T171S, T171S-N60A, and T171A-T223A, caused a considerable increase in L-asparagine affinity and a decrease in l-glutamine affinity as compared to the wild-type and other mutants. Then, molecular dynamics simulation and MM/GBSA free energy were applied to assess the stability of protein structure and its interaction with ligands. The three mutated proteins, especially T171S-N60A, had higher stability and interactions with L-asparagine than l-glutamine in comparison with the wild-type. The YL-ASNase mutants could be introduced as appropriate therapeutic candidates that might cause lower side effects. However, the functional properties of these mutated enzymes need to be confirmed by genetic manipulation and in vitro and in vivo studies.


Subject(s)
Antineoplastic Agents , Yarrowia , Asparaginase/chemistry , Glutamine/chemistry , Molecular Docking Simulation , Asparagine/metabolism , Yarrowia/genetics , Yarrowia/metabolism , Molecular Dynamics Simulation , Antineoplastic Agents/chemistry
18.
IUBMB Life ; 76(8): 505-522, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38391119

ABSTRACT

The amide proteogenic amino acids, asparagine and glutamine, are two of the twenty amino acids used in translation by all known life. The aminoacyl-tRNA synthetases for asparagine and glutamine, asparaginyl-tRNA synthetase and glutaminyl tRNA synthetase, evolved after the split in the last universal common ancestor of modern organisms. Before that split, life used two-step indirect pathways to synthesize asparagine and glutamine on their cognate tRNAs to form the aminoacyl-tRNA used in translation. These two-step pathways were retained throughout much of the bacterial and archaeal domains of life and eukaryotic organelles. The indirect routes use non-discriminating aminoacyl-tRNA synthetases (non-discriminating aspartyl-tRNA synthetase and non-discriminating glutamyl-tRNA synthetase) to misaminoacylate the tRNA. The misaminoacylated tRNA formed is then transamidated into the amide aminoacyl-tRNA used in protein synthesis by tRNA-dependent amidotransferases (GatCAB and GatDE). The enzymes and tRNAs involved assemble into complexes known as transamidosomes to help maintain translational fidelity. These pathways have evolved to meet the varied cellular needs across a diverse set of organisms, leading to significant variation. In certain bacteria, the indirect pathways may provide a means to adapt to cellular stress by reducing the fidelity of protein synthesis. The retention of these indirect pathways versus acquisition of asparaginyl-tRNA synthetase and glutaminyl tRNA synthetase in lineages likely involves a complex interplay of the competing uses of glutamine and asparagine beyond translation, energetic costs, co-evolution between enzymes and tRNA, and involvement in stress response that await further investigation.


Subject(s)
Amino Acyl-tRNA Synthetases , Evolution, Molecular , Protein Biosynthesis , RNA, Transfer, Amino Acyl , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Amino Acyl/genetics , Asparagine/metabolism , Asparagine/genetics , Glutamine/metabolism , Bacteria/genetics , Bacteria/enzymology , Bacteria/metabolism , Archaea/genetics , Archaea/metabolism , Archaea/enzymology , Aspartate-tRNA Ligase/genetics , Aspartate-tRNA Ligase/metabolism , Amides/metabolism , Humans
19.
Compr Rev Food Sci Food Saf ; 23(1): e13260, 2024 01.
Article in English | MEDLINE | ID: mdl-38284574

ABSTRACT

Thermal processing techniques can lead to the formation of heat-induced toxic substances. Acrylamide is one contaminant that has received much scientific attention in recent years, and it is formed essentially during the Maillard reaction when foods rich in carbohydrates, particularly reducing sugars (glucose, fructose), and certain free amino acids, especially asparagine (ASN), are processed at high temperatures (>120°C). The highly variable free ASN concentration in raw materials makes it challenging for food businesses to keep acrylamide content below the European Commission benchmark levels, while avoiding flavor, color, and texture impacts on their products. Free ASN concentrations in crops are affected by environment, genotype, and soil fertilization, which can also influence protein content and amino acid composition. This review aims to provide an overview of free ASN and acrylamide quantification methods and mitigation strategies for acrylamide formation in foods, focusing on adding pulse flours to cereal-based snacks and bakery products. Overall, this review emphasizes the importance of these mitigation strategies in minimizing acrylamide formation in plant-based products and ensuring safer and healthier food options.


Subject(s)
Asparagine , Edible Grain , Asparagine/analysis , Asparagine/chemistry , Asparagine/metabolism , Edible Grain/chemistry , Acrylamide/analysis , Acrylamide/chemistry , Acrylamide/toxicity , Snacks , Carbohydrates/analysis , Carbohydrates/chemistry , Amino Acids/analysis
20.
Nat Commun ; 15(1): 937, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38297033

ABSTRACT

Malaria poses an enormous threat to human health. With ever increasing resistance to currently deployed drugs, breakthrough compounds with novel mechanisms of action are urgently needed. Here, we explore pyrimidine-based sulfonamides as a new low molecular weight inhibitor class with drug-like physical parameters and a synthetically accessible scaffold. We show that the exemplar, OSM-S-106, has potent activity against parasite cultures, low mammalian cell toxicity and low propensity for resistance development. In vitro evolution of resistance using a slow ramp-up approach pointed to the Plasmodium falciparum cytoplasmic asparaginyl-tRNA synthetase (PfAsnRS) as the target, consistent with our finding that OSM-S-106 inhibits protein translation and activates the amino acid starvation response. Targeted mass spectrometry confirms that OSM-S-106 is a pro-inhibitor and that inhibition of PfAsnRS occurs via enzyme-mediated production of an Asn-OSM-S-106 adduct. Human AsnRS is much less susceptible to this reaction hijacking mechanism. X-ray crystallographic studies of human AsnRS in complex with inhibitor adducts and docking of pro-inhibitors into a model of Asn-tRNA-bound PfAsnRS provide insights into the structure-activity relationship and the selectivity mechanism.


Subject(s)
Antimalarials , Aspartate-tRNA Ligase , Animals , Humans , Plasmodium falciparum/genetics , Asparagine/metabolism , Aspartate-tRNA Ligase/genetics , RNA, Transfer, Amino Acyl/metabolism , Antimalarials/pharmacology , Mammals/genetics
SELECTION OF CITATIONS
SEARCH DETAIL