Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.361
Filter
1.
Phytomedicine ; 133: 155948, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153276

ABSTRACT

BACKGROUND: The incidence of invasive fungal diseases (IFDs), represented by Candida albicans infection, is increasing year by year. However, clinically available antifungal drugs are very limited and encounter challenges such as limited efficacy, drug resistance, high toxicity, and exorbitant cost. Therefore, there is an urgent need for new antifungal drugs. PURPOSE: This study aims to find new antifungal compounds from plants, preferably those with good activity and low toxicity, and reveal their antifungal targets. METHODS: In vitro antifungal activities of compounds were investigated using broth microdilution method, spot assay, hyphal growth assay and biofilm formation assay. Synergistic effects were assessed using broth microdilution checkerboard technique. In vivo antifungal activities were evaluated using Galleria mellonella and murine candidiasis models. Cytotoxicity of compounds was investigated using Cell Counting Kit-8 (CCK-8). Discovery and validation of antifungal targets of compounds were conducted by using monoallelic knockout library of C. albicans, haploinsufficiency profiling (HIP), thermal shift assay (TSA), enzyme inhibitory effect assay, molecular docking, and in vitro and in vivo antifungal studies. RESULTS: 814 plant products were screened, among which petroselinic acid (PeAc) was found as an antifungal molecule. As a rare fatty acid isolated from coriander (Coriandrum sativum), carrot (Daucus carota) and other plants of the Apiaceae family, PeAc had not previously been found to have antifungal effects. In this study, PeAc was revealed to inhibit the growth of various pathogenic fungi, exhibited synergistic effects with fluconazole (FLC), inhibited the formation of C. albicans hyphae and biofilms, and showed antifungal effects in vivo. PeAc was less toxic to mammalian cells. Fructose-1,6-bisphosphate aldolase (Fba1p) was identified as a target of PeAc by using HIP, TSA, enzyme inhibitory effect assay and molecular docking methods. PeAc exerted antifungal effects more effectively on fba1Δ/FBA1 than wild-type (WT) strain both in vitro and in vivo. CONCLUSIONS: PeAc is an effective and low toxic antifungal compound. The target of PeAc is Fba1p. Fba1p is a promising target for antifungal drug development.


Subject(s)
Antifungal Agents , Candida albicans , Candidiasis , Fructose-Bisphosphate Aldolase , Microbial Sensitivity Tests , Molecular Docking Simulation , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Animals , Candida albicans/drug effects , Mice , Fructose-Bisphosphate Aldolase/metabolism , Candidiasis/drug therapy , Biofilms/drug effects , Drug Synergism , Hyphae/drug effects , Petroselinum/chemistry , Moths/drug effects , Disease Models, Animal
2.
Nat Metab ; 6(8): 1505-1528, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39134903

ABSTRACT

Lysine ß-hydroxybutyrylation (Kbhb) is a post-translational modification induced by the ketogenic diet (KD), a diet showing therapeutic effects on multiple human diseases. Little is known how cellular processes are regulated by Kbhb. Here we show that protein Kbhb is strongly affected by the KD through a multi-omics analysis of mouse livers. Using a small training dataset with known functions, we developed a bioinformatics method for the prediction of functionally important lysine modification sites (pFunK), which revealed functionally relevant Kbhb sites on various proteins, including aldolase B (ALDOB) Lys108. KD consumption or ß-hydroxybutyrate supplementation in hepatocellular carcinoma cells increases ALDOB Lys108bhb and inhibits the enzymatic activity of ALDOB. A Kbhb-mimicking mutation (p.Lys108Gln) attenuates ALDOB activity and its binding to substrate fructose-1,6-bisphosphate, inhibits mammalian target of rapamycin signalling and glycolysis, and markedly suppresses cancer cell proliferation. Our study reveals a critical role of Kbhb in regulating cancer cell metabolism and provides a generally applicable algorithm for predicting functionally important lysine modification sites.


Subject(s)
Diet, Ketogenic , Lysine , Protein Processing, Post-Translational , Lysine/metabolism , Animals , Mice , Humans , Fructose-Bisphosphate Aldolase/metabolism , 3-Hydroxybutyric Acid/metabolism , Liver Neoplasms/metabolism , Neoplasms/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Proliferation
3.
Med Oncol ; 41(9): 224, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120781

ABSTRACT

Aldolase enzymes, particularly ALDOA, ALDOB, and ALDOC, play a crucial role in the development and progression of cancer. While the aldolase family is mainly known for its involvement in the glycolysis pathway, these enzymes also have various pathological and physiological functions through distinct signaling pathways such as Wnt/ß-catenin, EGFR/MAPK, Akt, and HIF-1α. This has garnered increased attention in recent years and shed light on other sides of this enzyme. Potential therapeutic strategies targeting aldolases include using siRNA, inhibitors like naphthol AS-E phosphate and TX-2098, and natural compounds such as HDPS-4II and L-carnosine. Additionally, anticancer peptides derived from ALDOA, like P04, can potentially increase cancer cells' sensitivity to chemotherapy. Aldolases also affect cancer drug resistance by different approaches, making them good therapeutic targets. In this review, we extensively explore the role of aldolase enzymes in various types of cancers in proliferation, invasion, migration, and drug resistance; we also significantly explore the possible treatment considering aldolase function.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Fructose-Bisphosphate Aldolase/metabolism , Animals
4.
Mol Metab ; 87: 101984, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38972375

ABSTRACT

OBJECTIVE: Stable isotope studies have shown that hepatic de novo lipogenesis (DNL) plays an important role in the pathogenesis of intrahepatic lipid (IHL) deposition. Furthermore, previous research has demonstrated that fructose 1-phosphate (F1P) not only serves as a substrate for DNL, but also acts as a signalling metabolite that stimulates DNL from glucose. The aim of this study was to elucidate the mediators of F1P-stimulated DNL, with special focus on two key regulators of intrahepatic glucose metabolism, i.e., glucokinase regulatory protein (GKRP) and carbohydrate response element binding protein (ChREBP). METHODS: Aldolase B deficient mice (Aldob-/-), characterized by hepatocellular F1P accumulation, enhanced DNL, and hepatic steatosis, were either crossed with GKRP deficient mice (Gckr-/-) or treated with short hairpin RNAs directed against hepatic ChREBP. RESULTS: Aldob-/- mice showed higher rates of de novo palmitate synthesis from glucose when compared to wildtype mice (p < 0.001). Gckr knockout reduced de novo palmitate synthesis in Aldob-/- mice (p = 0.017), without affecting the hepatic mRNA expression of enzymes involved in DNL. In contrast, hepatic ChREBP knockdown normalized the hepatic mRNA expression levels of enzymes involved in DNL and reduced fractional DNL in Aldob-/- mice (p < 0.05). Of interest, despite downregulation of DNL in response to Gckr and ChREBP attenuation, no reduction in intrahepatic triglyceride levels was observed. CONCLUSIONS: Both GKRP and ChREBP mediate F1P-stimulated DNL in aldolase B deficient mice. Further studies are needed to unravel the role of GKRP and hepatic ChREBP in regulating IHL accumulation in aldolase B deficiency.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Fructose-Bisphosphate Aldolase , Lipogenesis , Liver , Mice, Knockout , Triglycerides , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Mice , Liver/metabolism , Triglycerides/metabolism , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Male , Mice, Inbred C57BL , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Glucose/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Carrier Proteins
5.
Commun Biol ; 7(1): 849, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992061

ABSTRACT

Hereditary fructose intolerance (HFI) is a painful and potentially lethal genetic disease caused by a mutation in aldolase B resulting in accumulation of fructose-1-phosphate (F1P). No cure exists for HFI and treatment is limited to avoid exposure to fructose and sugar. Using aldolase B deficient mice, here we identify a yet unrecognized metabolic event activated in HFI and associated with the progression of the disease. Besides the accumulation of F1P, here we show that the activation of the purine degradation pathway is a common feature in aldolase B deficient mice exposed to fructose. The purine degradation pathway is a metabolic route initiated by adenosine monophosphate deaminase 2 (AMPD2) that regulates overall energy balance. We demonstrate that very low amounts of fructose are sufficient to activate AMPD2 in these mice via a phosphate trap. While blocking AMPD2 do not impact F1P accumulation and the risk of hypoglycemia, its deletion in hepatocytes markedly improves the metabolic dysregulation induced by fructose and corrects fat and glycogen storage while significantly increasing the voluntary tolerance of these mice to fructose. In summary, we provide evidence for a critical pathway activated in HFI that could be targeted to improve the metabolic consequences associated with fructose consumption.


Subject(s)
AMP Deaminase , Fructose Intolerance , Fructose-Bisphosphate Aldolase , Fructose , Animals , Male , Mice , AMP Deaminase/genetics , AMP Deaminase/metabolism , Disease Models, Animal , Energy Metabolism/drug effects , Fructose/metabolism , Fructose Intolerance/metabolism , Fructose Intolerance/genetics , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Fructosephosphates/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/etiology , Liver Diseases/genetics , Mice, Inbred C57BL , Mice, Knockout
6.
Dig Dis Sci ; 69(9): 3290-3304, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39068380

ABSTRACT

OBJECTIVE: Gastric cancer (GC) is believed to be one of the most common digestive tract malignant tumors. However, mounting evidence indicates a link between the glycolysis and tumorigenesis, including gastric cancer. METHODS: Our research identified 5508 differently expressed mRNAs in gastric cancer. Then, the genes highly associated with tumorigenesis were identified through weighted correlation network analysis (WGCNA). Bioinformatics analysis observed that these hub genes were significantly linked to the regulation of cell cycle, drug metabolism, and glycolysis. Among these hub genes, there is a critical gene involved in glycolysis regulation, namely fructose-bisphosphate B (ALDOB). RESULTS: Analysis based on The Cancer Genome Atlas (TCGA) and three Gene Expression Omnibus (GEO) datasets revealed that ALDOB was significantly downregulated in GC compared with normal tissues. In addition, cell viability assay confirmed that ALDOB acted as a tumor suppressor. Finally, drug sensitivity analysis revealed that ALDOB increased the sensitivity of gastric cancer cells to most antitumor drugs, especially talazoparib, XAV939, and FTI-277. Our results showed that the expression of ALDOB was significantly lower in GC tissues than in normal tissues. And ALDOB significantly inhibited proliferation and migration, delayed glycolysis in GC cells. Consequently, our study suggests that ALDOB may be a potential target for the clinical treatment of gastric cancer.


Subject(s)
Fructose-Bisphosphate Aldolase , Glycolysis , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/drug therapy , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Glycolysis/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Disease Progression , Cell Proliferation/drug effects , Cell Movement/drug effects
7.
FEBS Lett ; 598(15): 1864-1876, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38997224

ABSTRACT

Fructose bisphosphate aldolases (FBAs) catalyze the reversible cleavage of fructose 1,6-bisphosphate into dihydroxyacetone phosphate and glyceraldehyde 3-phosphate. We analyzed two previously uncharacterized cytosolic Arabidopsis FBAs, AtFBA4 and AtFBA5. Based on a recent report, we examined the interaction of AtFBA4 with calmodulin (CaM)-like protein 11 (AtCML11). AtFBA4 did not bind AtCML11; however, we found that CaM bound AtFBA5 in a Ca2+-dependent manner with high specificity and affinity (KD ~ 190 nm) and enhanced its stability. AtFBA4 and AtFBA5 exhibited Michaelis-Menten kinetics with Km and Vmax values of 180 µm and 4.9 U·mg-1 for AtFBA4, and 6.0 µm and 0.30 U·mg-1 for AtFBA5, respectively. The flavonoid morin inhibited both isozymes. Our study suggests that Ca2+ signaling and flavanols may influence plant glycolysis/gluconeogenesis.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Calmodulin , Flavonoids , Arabidopsis/metabolism , Arabidopsis/enzymology , Arabidopsis/genetics , Calmodulin/metabolism , Calmodulin/chemistry , Flavonoids/metabolism , Flavonoids/pharmacology , Flavonoids/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/chemistry , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/chemistry , Fructose-Bisphosphate Aldolase/antagonists & inhibitors , Calcium/metabolism , Kinetics , Protein Binding , Flavones
8.
Theranostics ; 14(10): 3793-3809, 2024.
Article in English | MEDLINE | ID: mdl-38994031

ABSTRACT

Rationale: CD8+ T cells undergo a series of metabolic reprogramming processes during their activation and proliferation, including increased glycolysis, decreased aerobic oxidation of sugars, increased amino acid metabolism and increased protein synthesis. However, it is still unclear what factors regulate these metabolic reprogramming processes in CD8+ T cells in the tumor immune microenvironment. Methods: T cell chromobox protein 4 (CBX4) knock-out mice models were used to determine the role of CBX4 in CD8+ T cells on the tumor immune microenvironment and tumor progression. Flow cytometry, Cut-Tag qPCR, Chip-seq, immunoprecipitation, metabolite detection, lentivirus infection and adoptive T cells transfer were performed to explore the underlying mechanisms of CBX4 knock-out in promoting CD8+ T cell activation and inhibiting tumor growth. Results: We found that CBX4 expression was induced in tumor-infiltrating CD8+ T cells and inhibited CD8+ T cell function by regulating glucose metabolism in tumor tissue. Mechanistically, CBX4 increases the expression of the metabolism-associated molecule aldolase B (Aldob) through sumoylation of trans-acting transcription factor 1 (SP1) and Krüppel-like factor 3 (KLF3). In addition, Aldob inhibits glycolysis and ATP synthesis in T cells by reducing the phosphorylation of the serine/threonine protein kinase (Akt) and ultimately suppresses CD8+ T cell function. Significantly, knocking out CBX4 may improve the efficacy of anti-PD-1 therapy by enhancing the function of CD8+ T cells in the tumor microenvironment. Conclusion: CBX4 is involved in CD8+ T cell metabolic reprogramming and functional persistence in tumor tissues, and serves as an inhibitor in CD8+ T cells' glycolysis and effector function.


Subject(s)
CD8-Positive T-Lymphocytes , Glycolysis , Mice, Knockout , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Tumor Microenvironment/immunology , Cell Line, Tumor , Mice, Inbred C57BL , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Humans , Cellular Reprogramming
9.
Cell Rep ; 43(8): 114550, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39058593

ABSTRACT

Despite being the leading cause of lung cancer-related deaths, the underlying molecular mechanisms driving metastasis progression are still not fully understood. Transfer RNA-derived fragments (tRFs) have been implicated in various biological processes in cancer. However, the role of tRFs in lung adenocarcinoma (LUAD) remains unclear. Our study identified a tRF, tRF-Val-CAC-024, associated with the high-risk component of LUAD, through validation using 3 cohorts. Our findings demonstrated that tRF-Val-CAC-024 acts as an oncogene in LUAD. Mechanistically, tRF-Val-CAC-024 was revealed to bind to aldolase A (ALDOA) dependent on Q125/E224 and promote the oligomerization of ALDOA, resulting in increased enzyme activity and enhanced aerobic glycolysis in LUAD cells. Additionally, we provide preliminary evidence of its potential clinical value by investigating the therapeutic effects of tRF-Val-CAC-024 antagomir-loaded lipid nanoparticles (LNPs) in cell-line-derived xenograft models. These results could enhance our understanding of the regulatory mechanisms of tRFs in LUAD and provide a potential therapeutic target.


Subject(s)
Adenocarcinoma of Lung , Fructose-Bisphosphate Aldolase , Glycolysis , Lung Neoplasms , RNA, Transfer , Humans , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Animals , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , RNA, Transfer/metabolism , RNA, Transfer/genetics , Cell Line, Tumor , Female , Male , Mice, Nude , Neoplasm Metastasis , Protein Multimerization , Mice, Inbred BALB C
10.
Cell Host Microbe ; 32(8): 1365-1379.e10, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39059397

ABSTRACT

Peptostreptococcus stomatis (P. stomatis) is enriched in colorectal cancer (CRC), but its causality and translational implications in CRC are unknown. Here, we show that P. stomatis accelerates colonic tumorigenesis in ApcMin/+ and azoxymethane/dextran sodium sulfate (AOM-DSS) models by inducing cell proliferation, suppressing apoptosis, and impairing gut barrier function. P. stomatis adheres to CRC cells through its surface protein fructose-1,6-bisphosphate aldolase (FBA) that binds to the integrin α6/ß4 receptor on CRC cells, leading to the activation of ERBB2 and the downstream MEK-ERK-p90 cascade. Blockade of the FBA-integrin α6/ß4 abolishes ERBB2-mitogen-activated protein kinase (MAPK) activation and the protumorigenic effect of P. stomatis. P. stomatis-driven ERBB2 activation bypasses receptor tyrosine kinase (RTK) blockade by EGFR inhibitors (cetuximab, erlotinib), leading to drug resistance in xenograft and spontaneous CRC models of KRAS-wild-type CRC. P. stomatis also abrogates BRAF inhibitor (vemurafenib) efficacy in BRAFV600E-mutant CRC xenografts. Thus, we identify P. stomatis as an oncogenic bacterium and a contributory factor for non-responsiveness to RTK inhibitors in CRC.


Subject(s)
Carcinogenesis , Colorectal Neoplasms , Drug Resistance, Neoplasm , Peptostreptococcus , Receptor, ErbB-2 , Animals , Humans , Mice , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , /pharmacology
11.
Int J Mol Sci ; 25(14)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39062929

ABSTRACT

The fructose-1,6-bisphosphate aldolase (FBA) gene family exists in higher plants, with the genes of this family playing significant roles in plant growth and development, as well as response to abiotic stresses. However, systematic reports on the FBA gene family and its functions in cucumber are lacking. In this study, we identified five cucumber FBA genes, named CsFBA1-5, that are distributed randomly across chromosomes. Phylogenetic analyses involving these cucumber FBAs, alongside eight Arabidopsis FBA proteins and eight tomato FBA proteins, were conducted to assess their homology. The CsFBAs were grouped into two clades. We also analyzed the physicochemical properties, motif composition, and gene structure of the cucumber FBAs. This analysis highlighted differences in the physicochemical properties and revealed highly conserved domains within the CsFBA family. Additionally, to explore the evolutionary relationships of the CsFBA family further, we constructed comparative syntenic maps with Arabidopsis and tomato, which showed high homology but only one segmental duplication event within the cucumber genome. Expression profiles indicated that the CsFBA gene family is responsive to various abiotic stresses, including low temperature, heat, and salt. Taken together, the results of this study provide a theoretical foundation for understanding the evolution of and future research into the functional characterization of cucumber FBA genes during plant growth and development.


Subject(s)
Cucumis sativus , Fructose-Bisphosphate Aldolase , Gene Expression Regulation, Plant , Phylogeny , Stress, Physiological , Cucumis sativus/genetics , Cucumis sativus/enzymology , Cucumis sativus/growth & development , Stress, Physiological/genetics , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Genome, Plant , Arabidopsis/genetics , Solanum lycopersicum/genetics , Multigene Family , Gene Expression Profiling , Chromosomes, Plant/genetics , Synteny/genetics , Chromosome Mapping
12.
Structure ; 32(9): 1322-1326.e4, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39013461

ABSTRACT

Two structures of fructose 6-phosphate aldolase, the wild-type and an engineered variant containing five active-site mutations, have been solved by cryoelectron microscopy (cryo-EM). The engineered variant affords production of aldols from aryl substituted ketones and aldehydes. This structure was solved to a resolution of 3.1 Å and contains the critical iminium reaction intermediate trapped in the active site. This provides new information that rationalizes the acquired substrate scope and aids in formulating hypotheses of the chemical mechanism. A Tyr residue (Y131) is positioned for a role as catalytic acid/base during the aldol reaction and the different structures demonstrate mobility of this amino acid residue. Further engineering of this fructose 6-phosphate aldolase (FSA) variant, guided by this new structure, identified additional FSA variants that display improved carboligation activities with 2-hydroxyacetophenone and phenylacetaldehyde.


Subject(s)
Aldehydes , Catalytic Domain , Fructose-Bisphosphate Aldolase , Ketones , Protein Engineering , Aldehydes/chemistry , Aldehydes/metabolism , Ketones/chemistry , Ketones/metabolism , Fructose-Bisphosphate Aldolase/chemistry , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Models, Molecular , Cryoelectron Microscopy , Substrate Specificity , Imines/chemistry , Imines/metabolism , Protein Binding , Acetaldehyde/chemistry , Acetaldehyde/metabolism , Acetaldehyde/analogs & derivatives , Tyrosine/chemistry , Tyrosine/metabolism , Aldehyde-Lyases , Escherichia coli Proteins
13.
Biomolecules ; 14(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39062466

ABSTRACT

Dihydroxyacetone phosphate (DHAP)-dependent aldolases catalyze the aldol addition of DHAP to a variety of aldehydes and generate compounds with two stereocenters. This reaction is useful to synthesize chiral acyclic nucleosides, which constitute a well-known class of antiviral drugs currently used. In such compounds, the chirality of the aliphatic chain, which mimics the open pentose residue, is crucial for activity. In this work, three DHAP-dependent aldolases: fructose-1,6-biphosphate aldolase from rabbit muscle, rhanmulose-1-phosphate aldolase from Thermotoga maritima, and fuculose-1-phosphate aldolase from Escherichia coli, were used as biocatalysts. Aldehyde derivatives of thymine and cytosine were used as acceptor substrates, generating new acyclic nucleoside analogues containing two new stereocenters with conversion yields between 70% and 90%. Moreover, structural analyses by molecular docking were carried out to gain insights into the diasteromeric excess observed.


Subject(s)
Aldehyde-Lyases , Escherichia coli , Fructose-Bisphosphate Aldolase , Molecular Docking Simulation , Pyrimidine Nucleosides , Thermotoga maritima , Animals , Escherichia coli/enzymology , Pyrimidine Nucleosides/chemistry , Pyrimidine Nucleosides/chemical synthesis , Aldehyde-Lyases/metabolism , Aldehyde-Lyases/chemistry , Rabbits , Fructose-Bisphosphate Aldolase/chemistry , Fructose-Bisphosphate Aldolase/metabolism , Thermotoga maritima/enzymology , Dihydroxyacetone Phosphate/metabolism , Dihydroxyacetone Phosphate/chemistry , Stereoisomerism
14.
Int J Biol Macromol ; 275(Pt 1): 132885, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838894

ABSTRACT

Fructose 1,6-bisphosphate aldolase (FBA) is a pivotal enzyme, which plays a critical role in fixing CO2 through the process of in the Calvin cycle. In this study, a comprehensive exploration of the FBA family genes in moso bamboo (Phyllostachys edulis) was conducted by the bioinformatics and biological analyses. A total of nine FBA genes (PeFBA1-PeFBA9) were identified in the moso bamboo genome. The expression patterns of PeFBAs across diverse tissues of moso bamboo suggested that they have multifaceted functionality. Notably, PeFBA8 might play an important role in regulating photosynthetic carbon metabolism. Co-expression and cis-element analyses demonstrated that PeFBA8 was regulated by a photosynthetic regulatory transcription factor (PeGLK1), which was confirmed by yeast one-hybrid and dual-luciferase assays. In-planta gene editing analysis revealed that the edited PeFBA8 mutants displayed compromised photosynthetic functionality, characterized by reduced electron transport rate and impaired photosystem I, leading to decreased photosynthesis rate overall, compared to the unedited control. The recombinant protein of PeFBA8 from prokaryotic expression exhibited enzymatic catalytic function. The findings suggest that the expression of PeFBA8 can affect photosynthetic efficiency of moso bamboo leaves, which underlines the potential of leveraging PeFBA8's regulatory mechanism to breed bamboo varieties with enhanced carbon fixation capability.


Subject(s)
Carbon , Gene Expression Regulation, Plant , Photosynthesis , Photosynthesis/genetics , Carbon/metabolism , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Poaceae/genetics , Poaceae/metabolism , Phylogeny
15.
Aging Dis ; 15(5): 2271-2283, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38739943

ABSTRACT

Prior studies have emphasized a bioenergetic crisis in the retinal pigment epithelium (RPE) as a critical factor in the development of age-related macular degeneration (AMD). The isoforms Fructose-1,6-bisphosphate aldolase C (ALDOC) and pyruvate kinase M2 (PKM2) have been proposed to play a role in AMD pathogenesis. While PKM2 and ALDOC are crucial for aerobic glycolysis in the neural retina, they are not as essential for the RPE. In this study, we examined the expression and activity of PKM2 and ALDOC in both young and aged RPE cells, as well as in the retina and RPE tissue of mice, including an experimentally induced AMD mouse model. Our findings reveal an upregulation in PKM2 and ALDOC expression, accompanied by increased pyruvate kinase activity, in the aged and AMD mouse RPE. Conversely, there is a decrease in ALDOC expression but an increase in PKM2 expression and pyruvate kinase activity in the aged and AMD retina. Overall, our study indicates that aged and AMD RPE cells tend to favor aerobic glycolysis, while this tendency is diminished in the aged and AMD retina. These results underscore the significance of targeting PKM2 and ALDOC in the RPE as a promising therapeutic approach to address the bioenergetic crisis and prevent vision loss in AMD.


Subject(s)
Fructose-Bisphosphate Aldolase , Glycolysis , Macular Degeneration , Pyruvate Kinase , Retinal Pigment Epithelium , Animals , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Mice , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/enzymology , Aging/metabolism , Humans , Mice, Inbred C57BL , Disease Models, Animal
16.
Stem Cell Res ; 78: 103451, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38820866

ABSTRACT

Hereditary fructose intolerance (HFI) is an autosomal recessive metabolic disease associated with a mutation in the aldolase B gene on chromosome 9q31. In this study, we generated a human-induced pluripotent stem cell (hiPSC) line, FDCHi015-A, from peripheral blood mononuclear cells (PBMCs) of a patient carrying the compound heterozygous mutations c.360_364delCAAA and c.1013C > T in exons 4 and 9 of the ALDOB gene, respectively. The iPSCs with the confirmed patient-specific mutation demonstrate pluripotency markers expression, a normal karyotype, and the ability to differentiate into derivatives of three germ layers.


Subject(s)
Induced Pluripotent Stem Cells , Leukocytes, Mononuclear , Mutation , Humans , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/cytology , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Cell Line , Cell Differentiation , Male , Karyotype
17.
J Biol Chem ; 300(4): 107147, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38460940

ABSTRACT

Zinc is required for many critical processes, including intermediary metabolism. In Saccharomyces cerevisiae, the Zap1 activator regulates the transcription of ∼80 genes in response to Zn supply. Some Zap1-regulated genes are Zn transporters that maintain Zn homeostasis, while others mediate adaptive responses that enhance fitness. One adaptive response gene encodes the 2-cysteine peroxiredoxin Tsa1, which is critical to Zn-deficient (ZnD) growth. Depending on its redox state, Tsa1 can function as a peroxidase, a protein chaperone, or a regulatory redox sensor. In a screen for possible Tsa1 regulatory targets, we identified a mutation (cdc19S492A) that partially suppressed the tsa1Δ growth defect. The cdc19S492A mutation reduced activity of its protein product, pyruvate kinase isozyme 1 (Pyk1), implicating Tsa1 in adapting glycolysis to ZnD conditions. Glycolysis requires activity of the Zn-dependent enzyme fructose-bisphosphate aldolase 1, which was substantially decreased in ZnD cells. We hypothesized that in ZnD tsa1Δ cells, the loss of a compensatory Tsa1 regulatory function causes depletion of glycolytic intermediates and restricts dependent amino acid synthesis pathways, and that the decreased activity of Pyk1S492A counteracted this depletion by slowing the irreversible conversion of phosphoenolpyruvate to pyruvate. In support of this model, supplementing ZnD tsa1Δ cells with aromatic amino acids improved their growth. Phosphoenolpyruvate supplementation, in contrast, had a much greater effect on growth rate of WT and tsa1Δ ZnD cells, indicating that inefficient glycolysis is a major factor limiting yeast growth. Surprisingly however, this restriction was not primarily due to low fructose-bisphosphate aldolase 1 activity, but instead occurs earlier in glycolysis.


Subject(s)
Glycolysis , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Zinc , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Zinc/metabolism , Fructose-Bisphosphate Aldolase/metabolism , Fructose-Bisphosphate Aldolase/genetics , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Gene Expression Regulation, Fungal , Peroxidases/metabolism , Peroxidases/genetics , Mutation
18.
Funct Integr Genomics ; 24(2): 53, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38453820

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the malignancies with the worst prognosis worldwide, in the occurrence and development of which glycolysis plays a central role. This study uncovered a mechanism by which ZNF692 regulates ALDOA-dependent glycolysis in HCC cells. RT-qPCR and western blotting were used to detect the expression of ZNF692, KAT5, and ALDOA in HCC cell lines and a normal liver cell line. The influences of transfection-induced alterations in the expression of ZNF692, KAT5, and ALDOA on the functions of HepG2 cells were detected by performing MTT, flow cytometry, Transwell, cell scratch, and colony formation assays, and the levels of glucose and lactate were determined using assay kits. ChIP and luciferase reporter assays were conducted to validate the binding of ZNF692 to the KAT5 promoter, and co-IP assays to detect the interaction between KAT5 and ALDOA and the acetylation of ALDOA. ZNF692, KAT5, and ALDOA were highly expressed in human HCC samples and cell lines, and their expression levels were positively correlated in HCC. ZNF692, ALDOA, or KAT5 knockdown inhibited glycolysis, proliferation, invasion, and migration and promoted apoptosis in HepG2 cells. ZNF692 bound to the KAT5 promoter and promoted its activity. ALDOA acetylation levels were elevated in HCC cell lines. KAT5 bound to ALDOA and catalyzed ALDOA acetylation. ALDOA or KAT5 overexpression in the same time of ZNF692 knockdown, compared to ZNF692 knockdown only, stimulated glycolysis, proliferation, invasion, and migration and reduced apoptosis in HepG2 cells. ZNF692 promotes the acetylation modification and protein expression of ALDOA by catalyzing KAT5 transcription, thereby accelerating glycolysis to drive HCC cell development.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Cell Line, Tumor , Hep G2 Cells , Glycolysis , Cell Proliferation , Gene Expression Regulation, Neoplastic , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism
19.
Sci Rep ; 14(1): 6488, 2024 03 18.
Article in English | MEDLINE | ID: mdl-38499636

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract and a leading cause of cancer-related death worldwide. Since many CRC patients are diagnosed already in the advanced stage, and traditional chemoradiotherapy is prone to drug resistance, it is important to find new therapeutic targets. In this study, the expression levels of ALDOA and p-AKT were detected in cancer tissues and paired normal tissues, and it was found that they were significantly increased in CRC tissues, and their high expression indicated poor prognosis. Moreover, a positive correlation between the expression of ALDOA and p-AKT was found in CRC tissues and paired normal tissues. In addition, the Kaplan-Meier analysis revealed that the group with both negative of ALDOA/p-AKT expression had longer five-year survival rates compared with the other group. Besides, the group with both high expression of ALDOA/p-AKT had a worse prognosis compared with the other group. Based on the expression of ALDOA and p-AKT in tumor tissues, we can effectively distinguish tumor tissues from normal tissues through cluster analysis. Furthermore, we constructed nomograms to predict 3-year and 5-year overall survival, showing that the expression of ALDOA/p-AKT plays a crucial role in predicting the prognosis of CRC patients. Therefore, ALDOA/p-AKT may act as a crucial role in CRC, which may provide new horizons for targeted therapies for CRC.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Prognosis , Kaplan-Meier Estimate , Colorectal Neoplasms/metabolism , Fructose-Bisphosphate Aldolase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL