Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 716
1.
Gen Comp Endocrinol ; 352: 114514, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38582175

Hormonal influence on hepatic function is a critical aspect of whole-body energy balance in vertebrates. Catecholamines and corticosteroids both influence hepatic energy balance via metabolite mobilization through glycogenolysis and gluconeogenesis. Elasmobranchs have a metabolic organization that appears to prioritize the mobilization of hepatic lipid as ketone bodies (e.g. 3-hydroxybutyrate [3-HB]), which adds complexity in determining the hormonal impact on hepatic energy balance in this taxon. Here, a liver perfusion was used to investigate catecholamine (epinephrine [E]) and corticosteroid (corticosterone [B] and 11-deoxycorticosterone [DOC]) effects on the regulation of hepatic glucose and 3-HB balance in the North Pacific Spiny dogfish, Squalus suckleyi. Further, hepatic enzyme activity involved in ketogenesis (3-hydroxybutyrate dehydrogenase), glycogenolysis (glycogen phosphorylase), and gluconeogenesis (phosphoenolpyruvate carboxykinase) were assessed in perfused liver tissue following hormonal application to discern effects on hepatic energy flux. mRNA transcript abundance key transporters of glucose (glut1 and glut4) and ketones (mct1 and mct2) and glucocorticoid function (gr, pepck, fkbp5, and 11ßhsd2) were also measured to investigate putative cellular components involved in hepatic responses. There were no changes in the arterial-venous difference of either metabolite in all hormone perfusions. However, perfusion with DOC increased gr transcript abundance and decreased flow rate of perfusions, suggesting a regulatory role for this corticosteroid. Phosphoenolpyruvate carboxykinase activity increased following all hormone treatments, which may suggest gluconeogenic function; E also increased 3-hydroxybutyrate dehydrogenase activity, suggesting a function in ketogenesis, and decreased pepck and fkbp5 transcript abundance, potentially showing some metabolic regulation. Overall, we demonstrate hormonal control of hepatic energy balance using liver perfusions at various levels of biological organization in an elasmobranch.


Squalus acanthias , Squalus , Animals , Glucose/metabolism , Squalus/metabolism , Squalus acanthias/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Phosphoenolpyruvate/metabolism , Liver/metabolism , 3-Hydroxybutyric Acid/pharmacology , 3-Hydroxybutyric Acid/metabolism , Ketone Bodies/metabolism , Gluconeogenesis , Hormones/metabolism , Adrenal Cortex Hormones/metabolism
2.
Aging (Albany NY) ; 15(22): 13384-13410, 2023 11 27.
Article En | MEDLINE | ID: mdl-38015723

A ketogenic diet (KD) and ß-hydroxybutyrate (ßOHB) have been widely reported as effective therapies for metabolic diseases. ß-Hydroxybutyrate dehydrogenase 1 (BDH1) is the rate-limiting enzyme in ketone metabolism. In this study, we examined the BDH1-mediated ßOHB metabolic pathway in the pathogenesis of diabetic kidney disease (DKD). We found that BDH1 is downregulated in the kidneys in DKD mouse models, patients with diabetes, and high glucose- or palmitic acid-induced human renal tubular epithelial (HK-2) cells. BDH1 overexpression or ßOHB treatment protects HK-2 cells from glucotoxicity and lipotoxicity by inhibiting reactive oxygen species overproduction. Mechanistically, BDH1-mediated ßOHB metabolism activates NRF2 by enhancing the metabolic flux of ßOHB-acetoacetate-succinate-fumarate. Moreover, in vivo studies showed that adeno-associated virus 9-mediated BDH1 renal expression successfully reverses fibrosis, inflammation, and apoptosis in the kidneys of C57 BKS db/db mice. Either ßOHB supplementation or KD feeding could elevate the renal expression of BDH1 and reverse the progression of DKD. Our results revealed a BDH1-mediated molecular mechanism in the pathogenesis of DKD and identified BDH1 as a potential therapeutic target for DKD.


Diabetes Mellitus , Diabetic Nephropathies , Animals , Humans , Mice , 3-Hydroxybutyric Acid/pharmacology , Antioxidants/therapeutic use , Diabetic Nephropathies/metabolism , Kidney/pathology , NF-E2-Related Factor 2/genetics , Hydroxybutyrate Dehydrogenase/metabolism
3.
Thromb Haemost ; 123(10): 1003-1015, 2023 Oct.
Article En | MEDLINE | ID: mdl-37399841

BACKGROUND: We aimed to investigate the role and mechanism of ß-hydroxybutyrate dehydrogenase 1 (Bdh1) in regulating macrophage oxidative stress in diabetes-induced atherosclerosis (AS). METHODS: We performed immunohistochemical analysis of femoral artery sections to determine differences in Bdh1 expression between normal participants, AS patients, and patients with diabetes-induced AS. Diabetic Apoe-/- mice and high-glucose (HG)-treated Raw264.7 macrophages were used to replicate the diabetes-induced AS model. The role of Bdh1 in this disease model was determined by adeno-associated virus (AAV)-mediated overexpression of Bdh1 or overexpression or silencing of Bdh1. RESULTS: We observed reduced expression of Bdh1 in patients with diabetes-induced AS, HG-treated macrophages, and diabetic Apoe-/- mice. AAV-mediated Bdh1 overexpression attenuated aortic plaque formation in diabetic Apoe-/- mice. Silencing of Bdh1 resulted in increased reactive oxygen species (ROS) production and an inflammatory response in macrophages, which were reversed by the ROS scavenger N-acetylcysteine. Overexpression of Bdh1 protected Raw264.7 cells from HG-induced cytotoxicity by inhibiting ROS overproduction. In addition, Bdh1 induced oxidative stress through nuclear factor erythroid-related factor 2 (Nrf2) activation by fumarate acid. CONCLUSION: Bdh1 attenuates AS in Apoe-/- mice with type 2 diabetes, accelerates lipid degradation, and reduces lipid levels by promoting ketone body metabolism. Moreover, it activates the Nrf2 pathway of Raw264.7 by regulating the metabolic flux of fumarate, which inhibits oxidative stress and leads to a decrease in ROS and inflammatory factor production.


Atherosclerosis , Diabetes Mellitus, Type 2 , Humans , Mice , Animals , Hydroxybutyrate Dehydrogenase/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Reactive Oxygen Species , Mice, Knockout, ApoE , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Oxidative Stress , Apolipoproteins E , Fumarates , Lipids
4.
Appl Environ Microbiol ; 89(6): e0036623, 2023 06 28.
Article En | MEDLINE | ID: mdl-37255440

Ketone bodies, including acetoacetate, 3-hydroxybutyrate, and acetone, are produced in the liver of animals during glucose starvation. Enzymes for the metabolism of (R)-3-hydroxybutyrate have been extensively studied, but little is known about the metabolism of its enantiomer (S)-3-hydroxybutyrate. Here, we report the characterization of a novel pathway for the degradation of (S)-3-hydroxybutyrate in anaerobic bacteria. We identify and characterize a stereospecific (S)-3-hydroxylbutyrate dehydrogenase (3SHBDH) from Desulfotomaculum ruminis, which catalyzes the reversible NAD(P)H-dependent reduction of acetoacetate to form (S)-3-hydroxybutyrate. 3SHBDH also catalyzes oxidation of d-threonine (2R, 3S) and l-allo-threonine (2S, 3S), consistent with its specificity for ß-(3S)-hydroxy acids. Isothermal calorimetry experiments support a sequential mechanism involving binding of NADH prior to (S)-3-hydroxybutyrate. Homologs of 3SHBDH are present in anaerobic fermenting and sulfite-reducing bacteria, and experiments with Clostridium pasteurianum showed that 3SHBDH, acetate CoA-transferase (YdiF), and (S)-3-hydroxybutyryl-CoA dehydrogenase (Hbd) are involved together in the degradation of (S)-3-hydroxybutyrate as a carbon and energy source for growth. (S)-3-hydroxybutyrate is a human metabolic marker and a chiral precursor for chemical synthesis, suggesting potential applications of 3SHBDH in diagnostics or the chemicals industry. IMPORTANCE (R)-3-hydroxybutyrate is well studied as a component of ketone bodies produced by the liver and of bacterial polyesters. However, the biochemistry of its enantiomer (S)-3-hydroxybutyrate is poorly understood. This study describes the identification and characterization of a stereospecific (S)-3-hydroxylbutyrate dehydrogenase and its function in a metabolic pathway for the degradation of (S)-3-hydroxybutyrate as a carbon and energy source in anaerobic bacteria. (S)-3-hydroxybutyrate is a mammalian metabolic marker and a precursor for chemical synthesis and bioplastics, suggesting potential applications of these enzymes in diagnostics and biotechnology.


Acetoacetates , Bacteria, Anaerobic , Animals , Humans , 3-Hydroxybutyric Acid , Bacteria, Anaerobic/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Hydroxybutyrates/metabolism , Ketone Bodies/metabolism , 3-Hydroxyacyl-CoA Dehydrogenase , Bacteria/metabolism , Carbon , Threonine , Mammals
5.
J Alzheimers Dis ; 92(2): 411-424, 2023.
Article En | MEDLINE | ID: mdl-36776048

BACKGROUND: The complex and not yet fully understood etiology of Alzheimer's disease (AD) shows important proteopathic signs which are unlikely to be linked to a single protein. However, protein subsets from deep proteomic datasets can be useful in stratifying patient risk, identifying stage dependent disease markers, and suggesting possible disease mechanisms. OBJECTIVE: The objective was to identify protein subsets that best classify subjects into control, asymptomatic Alzheimer's disease (AsymAD), and AD. METHODS: Data comprised 6 cohorts; 620 subjects; 3,334 proteins. Brain tissue-derived predictive protein subsets for classifying AD, AsymAD, or control were identified and validated with label-free quantification and machine learning. RESULTS: A 29-protein subset accurately classified AD (AUC = 0.94). However, an 88-protein subset best predicted AsymAD (AUC = 0.92) or Control (AUC = 0.92) from AD (AUC = 0.98). AD versus Control: APP, DHX15, NRXN1, PBXIP1, RABEP1, STOM, and VGF. AD versus AsymAD: ALDH1A1, BDH2, C4A, FABP7, GABBR2, GNAI3, PBXIP1, and PRKAR1B. AsymAD versus Control: APP, C4A, DMXL1, EXOC2, PITPNB, RABEP1, and VGF. Additional predictors: DNAJA3, PTBP2, SLC30A9, VAT1L, CROCC, PNP, SNCB, ENPP6, HAPLN2, PSMD4, and CMAS. CONCLUSION: Biomarkers were dynamically separable across disease stages. Predictive proteins were significantly enriched to sugar metabolism.


Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Proteomics , Brain/metabolism , Machine Learning , Sugars/metabolism , HSP40 Heat-Shock Proteins/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Proteins/metabolism
6.
FASEB J ; 36(11): e22626, 2022 11.
Article En | MEDLINE | ID: mdl-36305462

PTEN (phosphatase and tensin homolog deleted on chromosome 10) is one of the most frequently mutated/deleted tumor suppressor genes in many human cancers. Ursolic acid (UA) is a natural triterpenoid possessing antioxidant, anti-inflammatory, and anticancer effects. However, how PTEN impacts metabolic rewiring and how UA modifies PTEN-driven metabolic and epigenetic reprogramming in prostate cancer (PCa) remains unknown. In the current study, we found that UA protects against PTEN knockout (KO)-induced tumorigenesis at different stages of PCa. Epigenomic CpG methyl-seq revealed UA attenuated PTEN KO-induced differentially methylated regions (DMRs) profiles. Transcriptomic RNA-seq showed UA abrogated PTEN KO-induced differentially expressed genes (DEGs) of PCa-related oncogenes' Has3, Cfh, and Msx1 overexpression, indicating UA plays a crucial role in PTEN KO-mediated gene regulation and its potential consequences on cancer interception. Association analysis of DEGs and DMRs identified that the mRNA expression of tumor suppressor gene BDH2, and oncogenes Ephas, Isg15, and Nos2 were correlated with the promoter CpG methylation status in the early-stage comparison groups indicating UA could regulate the oncogenes or tumor suppressor genes by modulating their promoter methylation at an early stage of prostate tumorigenesis. The metabolomic study showed UA attenuated PTEN KO-regulated cancer-associated metabolisms like purine metabolism/metabolites correlating with RNAseq findings, glycolysis/gluconeogenesis metabolism, as well as epigenetic-related metabolites pyruvate and lactate indicating UA plays a critical role in PTEN KO-mediated metabolic and epigenetic reprogramming and its consequences on cancer development. In this context, UA impacts metabolic rewiring causing epigenetic and transcriptomic reprogramming potentially contributing to the overall protection against prostate-specific PTEN KO-mediated PCa.


Prostatic Neoplasms , Triterpenes , Male , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Chemoprevention , Epigenesis, Genetic , Epigenomics , Hydroxybutyrate Dehydrogenase/genetics , Hydroxybutyrate Dehydrogenase/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/prevention & control , Prostatic Neoplasms/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Triterpenes/pharmacology , Mice, Knockout , Ursolic Acid
7.
Diabetes Obes Metab ; 24(11): 2263-2272, 2022 11.
Article En | MEDLINE | ID: mdl-35801343

AIM: To investigate cardiac signalling pathways connecting substrate utilization with left ventricular remodelling in a murine pressure overload model. METHODS: Cardiac hypertrophy was induced by transverse aortic constriction surgery in 20-week-old C57BL/6J mice treated with or without the sodium-glucose co-transporter 2 (SGLT2) inhibitor ertugliflozin (225 mg kg-1 chow diet) for 10 weeks. RESULTS: Ertugliflozin improved left ventricular function and reduced myocardial fibrosis. This occurred simultaneously with a fasting-like response characterized by improved glucose tolerance and increased ketone body concentrations. While cardiac insulin signalling was reduced in response to SGLT2 inhibition, AMP-activated protein kinase (AMPK) signalling was increased with induction of the fatty acid transporter cluster of differentiation 36 and phosphorylation of acetyl-CoA carboxylase (ACC). Further, enzymes responsible for ketone body catabolism (ß-hydroxybutyrate dehydrogenase, succinyl-CoA:3-oxoacid-CoA transferase and acetyl-CoA acetyltransferase 1) were induced by SGLT2 inhibition. Ertugliflozin led to more cardiac abundance of fatty acids, tricarboxylic acid cycle metabolites and ATP. Downstream mechanistic target of rapamycin (mTOR) pathway, relevant for protein synthesis, cardiac hypertrophy and adverse cardiac remodelling, was reduced by SGLT2 inhibition, with alleviation of endoplasmic reticulum (ER) stress and unfolded protein response (UPR) providing a potential mechanism for abundant reduced left ventricular apoptosis and fibrosis. CONCLUSION: SGLT2 inhibition reduced left ventricular fibrosis in a murine model of cardiac hypertrophy. Mechanistically, this was associated with reduced cardiac insulin and increased AMPK signalling as a potential mechanism for less cardiac mTOR activation with alleviation of downstream ER stress, UPR and apoptosis.


Insulins , Sodium-Glucose Transporter 2 Inhibitors , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic , Cardiomegaly/metabolism , Cardiomegaly/pathology , Coenzyme A-Transferases/metabolism , Endoplasmic Reticulum Stress , Fatty Acids/metabolism , Fibrosis , Glucose/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Keto Acids/metabolism , Ketones/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Sirolimus/metabolism , Sodium/metabolism , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/metabolism
8.
J Clin Invest ; 132(9)2022 05 02.
Article En | MEDLINE | ID: mdl-35499081

T follicular helper (Tfh) cells are a subset of CD4+ T cells that are essential in the pathogenesis of systemic lupus erythematosus (SLE). Notably, iron is required for activated CD4+ T lymphocytes to sustain high proliferation and metabolism. In this issue of the JCI, Gao et al. showed that CD4+ T cells from patients with SLE accumulated iron, augmenting their differentiation into Tfh cells and correlating with disease activity. Using human cells and murine models, the authors demonstrated that miR-21 was overexpressed in lupus T cells and inhibited 3-hydroxybutyrate dehydrogenase-2 (BDH2). The subsequent loss of BDH2 drove labile iron to accumulate in the cytoplasm and promoted TET enzyme activity, BCL6 gene demethylation, and Tfh cell differentiation. This work identifies a role for iron in CD4+ T cell biology and the development of pathogenic effectors in SLE. We await future investigations that could determine whether modulating iron levels could regulate Tfh cells in human health and disease.


Lupus Erythematosus, Systemic , T-Lymphocytes, Helper-Inducer , Animals , Humans , Hydroxybutyrate Dehydrogenase/metabolism , Iron/metabolism , Lupus Erythematosus, Systemic/genetics , Lymphocyte Activation , Mice , T Follicular Helper Cells
9.
Food Chem Toxicol ; 163: 112970, 2022 May.
Article En | MEDLINE | ID: mdl-35367536

The present study focused on the protective effects of melatonin against cisplatin-induced acute kidney injury in mice and its possible mechanism of action in relation to the major regulator of fatty acid oxidation (FAO), peroxidase proliferative receptor α (PPARα). The experiment consisted of the following four groups: vehicle control, cisplatin (15 mg/kg), cisplatin & melatonin (20 mg/kg/day), and melatonin (20 mg/kg/day). Concomitant administration of melatonin significantly ameliorated cisplatin-induced acute kidney injury in mice by decreasing serum levels of triglyceride, blood urea nitrogen and creatinine, reducing the number and size of lipid droplets in tubular epithelial cells, and decreasing the incidence of histopathological changes including tubular cell apoptosis. Moreover, melatonin administration protected kidney tissue by significantly upregulating the levels of PPARα reduced by cisplatin injection, resulting in increased FAO pathway-associated genes (PGC-1a, Acadm, Acat1, Acsm2, Acsm3, Bdh2, Echs and Pecr) as well as reducing protein levels of caspase-3, -9 and Bax. Melatonin not only partially modulated FAO via PPARα signaling, but also decreased cisplatin-induced apoptosis by inhibiting the caspase-3, -9 and Bax pathways. Our findings suggest that melatonin prevents cisplatin-induced acute kidney injury in mice, possibly by upregulating the expression of PPARα, resulting in enhanced FAO and anti-apoptotic properties.


Acute Kidney Injury , Melatonin , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Animals , Apoptosis , Caspase 3/metabolism , Cisplatin/metabolism , Cisplatin/toxicity , Fatty Acids/metabolism , Female , Humans , Hydroxybutyrate Dehydrogenase/metabolism , Hydroxybutyrate Dehydrogenase/pharmacology , Kidney , Male , Melatonin/pharmacology , Mice , PPAR alpha/genetics , PPAR alpha/metabolism , bcl-2-Associated X Protein/metabolism
10.
Gen Physiol Biophys ; 41(2): 115-122, 2022 Mar.
Article En | MEDLINE | ID: mdl-35416174

Cytoprotective autophagy induces tumor cell apoptosis or autophagic programmed cell death. Autophagy and apoptosis are implicated in the pathogenesis of lung cancer, especially lung adenocarcinoma. 3-Hydroxybutyrate dehydrogenase type 2 (BDH2), a rate-limiting catalyzer in the regulation of intracellular iron metabolism and siderophore biogenesis, has been shown to be a tumor suppressor through promotion of cell apoptosis and autophagy. However, the biological role of BDH2 on lung adenocarcinoma cell apoptosis and autophagy remains unclear. Data from Western blot and qRT-PCR showed that BDH2 was down-regulated in lung adenocarcinoma cells (A549, NCI-H1975, PC9) compared to normal human lung cells (BEAS-2B). Functional assays demonstrated that pcDNA-mediated over-expression of BDH2 reduced cell viability of lung adenocarcinoma cells, and repressed the proliferation. Cell apoptosis of lung adenocarcinoma was promoted by BDH2 over-expression with up-regulation of Bax and cleaved caspase-3. Over-expression of BDH2 reduced protein expression of p62 in lung adenocarcinoma cells, enhanced LC3 and Beclin-1. Phosphorylation of AKT and mTOR in lung adenocarcinoma cells were reduced by BDH2 over-expression. In conclusion, BDH2 functioned as a tumor suppressor in lung adenocarcinoma through promotion of Akt/mTOR-mediated cell apoptosis and autophagy.


Adenocarcinoma of Lung , Lung Neoplasms , Apoptosis , Autophagy , Cell Line, Tumor , Cell Proliferation , Humans , Hydroxybutyrate Dehydrogenase/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
Int J Biol Markers ; 37(2): 158-169, 2022 Jun.
Article En | MEDLINE | ID: mdl-35254116

BACKGROUND: Fragile histidine triad (FHIT) is a strong tumor suppressor gene, and cells deficient in FHIT are prone to acquiring cancer-promoting mutations. Due to its location, deletions within FHIT are common in cancer. Over 50% of cancers show loss of FHIT expression. However, to date, expression levels, gene regulatory networks, prognostic value, and target prediction of FHIT in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) have not been fully reported. Therefore, systematic analysis of FHIT expression, gene regulatory network, prognostic value, and targeted prediction in patients with LUAD and LUSC has important guiding significance, providing new therapeutic targets and strategies for clinical treatment of lung cancer to further improve the therapeutic effect of lung cancer. METHODS: Multiple free online databases were used for the abovementioned analysis in this study, including cBioPortal, TRRUST, Human Protein Atlas, GeneMANIA, GEPIA, Metascape, UALCAN, LinkedOmics, and TIMER. RESULTS: FHIT was upregulated in patients with LUAD, and downregulated in patients with LUSC. Genetic alterations of FHIT were found in patients with LUAD (7%), and LUSC (10%). The promoter methylation of FHIT was lower in patients with LUAD and LUSC. FHIT expression significantly correlated with LUSC pathological stages. Furthermore, patients with LUAD and LUSC having low FHIT expression levels had a longer survival than those having high FHIT expression levels. FHIT and its neighboring genes (the 50 most frequently altered neighboring genes of FHIT) functioned in the regulation of protein kinase and DNA binding in patients with LUAD, as well as cell channels and membrane potential in patients with LUSC. Gene ontology enrichment analysis revealed that the functions of FHIT and its neighboring genes are mainly related to disordered domain-specific binding, protein kinase binding, and ion gated channel activity in patients with LUAD, as well as calcium ion binding and intracellular ligand-gated ion channel activity in patients with LUSC. Transcription factor targets of FHIT and its neighboring genes in patients with lung cancer were found: USF1, SOX6, USF2, SIRT1, VHL, LEF1, EZH2, TP53, HDAC1, ESR1, EGR1, YY1, MYC, RELA, NFKB1, and E2F1 in LUAD; and HDAC1, DNMT1, and E2F1 in LUSC. We further explored the FHIT-associated kinase (PRKCQ, AURKB and ATM in LUAD as well as PLK3 in LUSC) and FHIT-associated miRNA targets (MIR-188, MIR-323, and MIR-518A-2 in LUAD). Furthermore, the following genes had the strongest correlation with FHIT expression in patients with lung cancer: NICN1, HEMK1, and BDH2 in LUAD, and ZMAT1, TTC21A, and NICN1 in LUSC. FHIT expression was positively associated with immune cell infiltration (B cell) in patients with LUAD, as well as B cell, CD8 + T, CD4 + T cells, macrophages, and dendritic cells in patients with LUSC. Nevertheless, FHIT expression was negatively associated with CD8 + T cells and neutrophils in patients with LUAD. CONCLUSIONS: The expression, gene regulatory network, prognostic value and targeted prediction of FHIT in patients with LUAD and LUSC were systematically analyzed and revealed in this study, thereby laying a foundation for further research on the role of FHIT in LUAD and LUSC occurrence. This study provides new LUAD and LUSC therapeutic targets and prognostic biomarkers as a reference for fundamental and clinical research.


Acid Anhydride Hydrolases , Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Neoplasm Proteins , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Hydroxybutyrate Dehydrogenase/genetics , Hydroxybutyrate Dehydrogenase/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prognosis
12.
J Biol Chem ; 298(3): 101708, 2022 03.
Article En | MEDLINE | ID: mdl-35150746

Early studies revealed that chicken embryos incubated with a rare analog of l-proline, 4-oxo-l-proline, showed increased levels of the metabolite 4-hydroxy-l-proline. In 1962, 4-oxo-l-proline reductase, an enzyme responsible for the reduction of 4-oxo-l-proline, was partially purified from rabbit kidneys and characterized biochemically. However, only recently was the molecular identity of this enzyme solved. Here, we report the purification from rat kidneys, identification, and biochemical characterization of 4-oxo-l-proline reductase. Following mass spectrometry analysis of the purified protein preparation, the previously annotated mammalian cytosolic type 2 (R)-ß-hydroxybutyrate dehydrogenase (BDH2) emerged as the only candidate for the reductase. We subsequently expressed rat and human BDH2 in Escherichia coli, then purified it, and showed that it catalyzed the reversible reduction of 4-oxo-l-proline to cis-4-hydroxy-l-proline via chromatographic and tandem mass spectrometry analysis. Specificity studies with an array of compounds carried out on both enzymes showed that 4-oxo-l-proline was the best substrate, and the human enzyme acted with 12,500-fold higher catalytic efficiency on 4-oxo-l-proline than on (R)-ß-hydroxybutyrate. In addition, human embryonic kidney 293T (HEK293T) cells efficiently metabolized 4-oxo-l-proline to cis-4-hydroxy-l-proline, whereas HEK293T BDH2 KO cells were incapable of producing cis-4-hydroxy-l-proline. Both WT and KO HEK293T cells also produced trans-4-hydroxy-l-proline in the presence of 4-oxo-l-proline, suggesting that the latter compound might interfere with the trans-4-hydroxy-l-proline breakdown in human cells. We conclude that BDH2 is a mammalian 4-oxo-l-proline reductase that converts 4-oxo-l-proline to cis-4-hydroxy-l-proline and not to trans-4-hydroxy-l-proline, as originally thought. We also hypothesize that this enzyme may be a potential source of cis-4-hydroxy-l-proline in mammalian tissues.


Amino Acid Oxidoreductases , Hydroxybutyrate Dehydrogenase , Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/metabolism , Animals , Chick Embryo , Escherichia coli/metabolism , HEK293 Cells , Humans , Hydroxybutyrate Dehydrogenase/chemistry , Hydroxybutyrate Dehydrogenase/metabolism , Hydroxyproline/chemistry , Hydroxyproline/metabolism , Mammals/metabolism , Proline/analogs & derivatives , Proline/metabolism , Rabbits , Rats
13.
Bioengineered ; 13(3): 5103-5112, 2022 03.
Article En | MEDLINE | ID: mdl-35164665

RAB27B is a member of Ras-like small GTPases that plays a role in endocytosis, exocytosis, and vesicle trafficking. We made an attempt to study the impacts of RAB27B on the proliferation and apoptosis of acute myeloid leukemia (AML) cells. The silencing of RAB27B was induced by siRNA for the detection of proliferation, cell cycle, and apoptosis, respectively by Cell Counting Kit-8 (CCK8), flow cytometry, and TUNEL. Related markers were also evaluated by Western blot analysis. The interaction between RAB27B and BDH2 was predicted by bioinformatics analysis and determined by immunoprecipitation. The gain of function of BDH2 was also detected by these functional assays. RAB27B exhibited high levels in AML cells, and RAB27B silencing led to reduced proliferation, increased cell cycle arrest and apoptosis levels. Then, the interaction between RAB27B and BDH2 was confirmed. Moreover, the effects of RAB27B inhibition on the proliferation, cell cycle arrest, and cell apoptosis were abolished after BDH2 overexpression. RAB27B inhibits proliferation and promotes apoptosis of leukemic cells by interacting with BDH2. Targeting RAB27B might be an effective method for the treatment of AML.


Leukemia, Myeloid, Acute , Apoptosis/genetics , Butyrates , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Hydroxybutyrate Dehydrogenase/genetics , Hydroxybutyrate Dehydrogenase/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Oxidoreductases/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
14.
Biochemistry ; 60(27): 2186-2194, 2021 07 13.
Article En | MEDLINE | ID: mdl-34190541

(R)-3-Hydroxybutyrate dehydrogenase (HBDH) catalyzes the NADH-dependent reduction of 3-oxocarboxylates to (R)-3-hydroxycarboxylates. The active sites of a pair of cold- and warm-adapted HBDHs are identical except for a single residue, yet kinetics evaluated at -5, 0, and 5 °C show a much higher steady-state rate constant (kcat) for the cold-adapted than for the warm-adapted HBDH. Intriguingly, single-turnover rate constants (kSTO) are strikingly similar between the two orthologues. Psychrophilic HBDH primary deuterium kinetic isotope effects on kcat (Dkcat) and kSTO (DkSTO) decrease at lower temperatures, suggesting more efficient hydride transfer relative to other steps as the temperature decreases. However, mesophilic HBDH Dkcat and DkSTO are generally temperature-independent. The DkSTO data allowed calculation of intrinsic primary deuterium kinetic isotope effects. Intrinsic isotope effects of 4.2 and 3.9 for cold- and warm-adapted HBDH, respectively, at 5 °C, supported by quantum mechanics/molecular mechanics calculations, point to a late transition state for both orthologues. Conversely, intrinsic isotope effects of 5.7 and 3.1 for cold- and warm-adapted HBDH, respectively, at -5 °C indicate the transition state becomes nearly symmetric for the psychrophilic enzyme, but more asymmetric for the mesophilic enzyme. His-to-Asn and Asn-to-His mutations in the psychrophilic and mesophilic HBDH active sites, respectively, swap the single active-site position where these orthologues diverge. At 5 °C, the His-to-Asn mutation in psychrophilic HBDH decreases Dkcat to 3.1, suggesting a decrease in transition-state symmetry, while the His-to-Asn mutation in mesophilic HBDH increases Dkcat to 4.4, indicating an increase in transition-state symmetry. Hence, temperature adaptation and a single divergent active-site residue may influence transition-state geometry in HBDHs.


Bacterial Proteins/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Psychrobacter/enzymology , Bacterial Proteins/chemistry , Catalytic Domain , Cold Temperature , Hydroxybutyrate Dehydrogenase/chemistry , Kinetics , Models, Molecular , Psychrobacter/chemistry , Psychrobacter/metabolism
15.
Mol Metab ; 53: 101269, 2021 11.
Article En | MEDLINE | ID: mdl-34116232

OBJECTIVE: Throughout the last decade, interest has intensified in intermittent fasting, ketogenic diets, and exogenous ketone therapies as prospective health-promoting, therapeutic, and performance-enhancing agents. However, the regulatory roles of ketogenesis and ketone metabolism on liver homeostasis remain unclear. Therefore, we sought to develop a better understanding of the metabolic consequences of hepatic ketone body metabolism by focusing on the redox-dependent interconversion of acetoacetate (AcAc) and D-ß-hydroxybutyrate (D-ßOHB). METHODS: Using targeted and isotope tracing high-resolution liquid chromatography-mass spectrometry, dual stable isotope tracer nuclear magnetic resonance spectroscopy-based metabolic flux modeling, and complementary physiological approaches in novel cell type-specific knockout mice, we quantified the roles of hepatocyte D-ß-hydroxybutyrate dehydrogenase (BDH1), a mitochondrial enzyme required for NAD+/NADH-dependent oxidation/reduction of ketone bodies. RESULTS: Exogenously administered AcAc is reduced to D-ßOHB, which increases hepatic NAD+/NADH ratio and reflects hepatic BDH1 activity. Livers of hepatocyte-specific BDH1-deficient mice did not produce D-ßOHB, but owing to extrahepatic BDH1, these mice nonetheless remained capable of AcAc/D-ßOHB interconversion. Compared to littermate controls, hepatocyte-specific BDH1 deficient mice exhibited diminished liver tricarboxylic acid (TCA) cycle flux and impaired gluconeogenesis, but normal hepatic energy charge overall. Glycemic recovery after acute insulin challenge was impaired in knockout mice, but they were not more susceptible to starvation-induced hypoglycemia. CONCLUSIONS: Ketone bodies influence liver homeostasis. While liver BDH1 is not required for whole body equilibration of AcAc and D-ßOHB, loss of the ability to interconvert these ketone bodies in hepatocytes results in impaired TCA cycle flux and glucose production. Therefore, through oxidation/reduction of ketone bodies, BDH1 is a significant contributor to hepatic mitochondrial redox, liver physiology, and organism-wide ketone body homeostasis.


Glucose/biosynthesis , Hepatocytes/metabolism , Hydroxybutyrate Dehydrogenase/metabolism , Ketones/metabolism , Animals , Citric Acid Cycle , Female , Hydroxybutyrate Dehydrogenase/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
J Exp Clin Cancer Res ; 39(1): 123, 2020 Jun 30.
Article En | MEDLINE | ID: mdl-32605589

BACKGROUND: 3-Hydroxy butyrate dehydrogenase 2 (BDH2) is a short-chain dehydrogenase/reductase family member that plays a key role in the development and pathogenesis of human cancers. However, the role of BDH2 in gastric cancer (GC) remains largely unclear. Our study aimed to ascertain the regulatory mechanisms of BDH2 in GC, which could be used to develop new therapeutic strategies. METHODS: Western blotting, immunohistochemistry, and RT-PCR were used to investigate the expression of BDH2 in GC specimens and cell lines. Its correlation with the clinicopathological characteristics and prognosis of GC patients was analysed. Functional assays, such as CCK-8 and TUNEL assays, transmission electron microscopy, and an in vivo tumour growth assay, were performed to examine the proliferation, apoptosis, and autophagy of GC cells. Related molecular mechanisms were clarified by luciferase reporter, coimmunoprecipitation, and ubiquitination assays. RESULTS: BDH2 was markedly downregulated in GC tissues and cells, and the low expression of BDH2 was associated with poor survival of GC patients. Functionally, BDH2 overexpression significantly induced apoptosis and autophagy in vitro and in vivo. Mechanistically, BDH2 promoted Keap1 interaction with Nrf2 to increase the ubiquitination level of Nrf2. Ubiquitination/degradation of Nrf2 inhibited the activity of ARE to increase accumulation of reactive oxygen species (ROS), thereby inhibiting the phosphorylation levels of AktSer473 and mTORSer2448. CONCLUSIONS: Our study indicates that BDH2 is an important tumour suppressor in GC. BDH2 regulates intracellular ROS levels to mediate the PI3K/Akt/mTOR pathway through Keap1/Nrf2/ARE signalling, thereby inhibiting the growth of GC.


Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Hydroxybutyrate Dehydrogenase/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Stomach Neoplasms/pathology , Ubiquitin/metabolism , Animals , Apoptosis , Autophagy , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Female , Humans , Hydroxybutyrate Dehydrogenase/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , NF-E2-Related Factor 2/genetics , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Rate , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
17.
Dig Liver Dis ; 52(10): 1076-1079, 2020 10.
Article En | MEDLINE | ID: mdl-32507692

Objective: To explore the clinical characteristics of Coronavirus Disease (COVID-19) patients with gastrointestinal symptoms. Methods: The clinical data of 164 COVID-19 patients with gastrointestinal symptoms were extracted and analysed retrospectively. Results: In total, 505 COVID-19 patients were divided into two groups: those with gastrointestinal symptoms (G group) and those without gastrointestinal symptoms (NG group). Common gastrointestinal symptoms included inappetence, diarrhoea, nausea, abdominal pain, and vomiting. Significantly higher proportions of patients with fever, dizziness, myalgia, and fatigue were noted in group G than in group NG. Compared with patients without fever, there was a significant difference between G group and NG group in moderate fever or above, while there was no significant difference between the two groups in low fever. The laboratory results showed that patients in the G group had significantly higher C-reactive protein, lactate dehydrogenase, and α-hydroxybutyrate dehydrogenase levels than those in the NG group. Moreover, the proportion of patients with severe pneumonia was significantly higher in the G group than in the NG group. Conclusion: In Wuhan, the proportion of COVID-19 patients who experience gastrointestinal symptoms is relatively high. Patients who experience gastrointestinal symptoms are more likely to suffer from severe pneumonia, which may help clinicians identify patients at high risk of COVID-19 and thus reduce the incidence of this condition.


Abdominal Pain/physiopathology , Anorexia/physiopathology , Coronavirus Infections/physiopathology , Diarrhea/physiopathology , Nausea/physiopathology , Pneumonia, Viral/physiopathology , Vomiting/physiopathology , Abdominal Pain/etiology , Abdominal Pain/metabolism , Adult , Aged , Anorexia/etiology , Anorexia/metabolism , Betacoronavirus , C-Reactive Protein/metabolism , COVID-19 , Case-Control Studies , Coronavirus Infections/complications , Coronavirus Infections/metabolism , Diarrhea/etiology , Diarrhea/metabolism , Dizziness/etiology , Dizziness/physiopathology , Fatigue/etiology , Fatigue/physiopathology , Female , Fever/etiology , Fever/physiopathology , Humans , Hydroxybutyrate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Middle Aged , Myalgia/etiology , Myalgia/physiopathology , Nausea/etiology , Nausea/metabolism , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/metabolism , Retrospective Studies , SARS-CoV-2 , Severity of Illness Index , Vomiting/etiology , Vomiting/metabolism
18.
PLoS Med ; 17(6): e1003130, 2020 06.
Article En | MEDLINE | ID: mdl-32544155

BACKGROUND: As of April 18, 2020, over 2,000,000 patients had been diagnosed with coronavirus disease-2019 (COVID-19) globally, and more than 140,000 deaths had been reported. The clinical and epidemiological characteristics of adult patients have been documented recently. However, information on pediatric patients is limited. We describe the clinical and epidemiological characteristics of pediatric patients to provide valuable insight into the early diagnosis and assessment of COVID-19 in children. METHODS AND FINDINGS: This retrospective, observational study involves a case series performed at 4 hospitals in West China. Thirty-four pediatric patients with COVID-19 were included from January 27 to February 23, 2020. The final follow-up visit was completed by March 16, 2020. Clinical and epidemiological characteristics were analyzed on the basis of demographic data, medical history, laboratory tests, radiological findings, and treatment information. Data analysis was performed for 34 pediatrics patients with COVID-19 aged from 1 to 144 months (median 33.00, interquartile range 10.00-94.25), among whom 14 males (41%) were included. All the patients in the current study presented mild (18%) or moderate (82%) forms of COVID-19. A total of 48% of patients were noted to be without a history of exposure to an identified source. Mixed infections of other respiratory pathogens were reported in 16 patients (47%). Comorbidities were reported in 6 patients (18%). The most common initial symptoms were fever (76%) and cough (62%). Expectoration (21%), vomiting (12%), and diarrhea (12%) were also reported in a considerable portion of cases. A substantial increase was detected in serum amyloid A for 17 patients (among 20 patients with available data; 85%) and in high-sensitivity C-reactive protein for 17 patients (among 29 patients with available data; 59%), whereas a decrease in prealbumin was noticed in 25 patients (among 32 patients with available data; 78%). In addition, significant increases in the levels of lactate dehydrogenase and α-hydroxybutyrate dehydrogenase were detected in 28 patients (among 34 patients with available data; 82%) and 25 patients (among 34 patients with available data; 74%), respectively. Patchy lesions in lobules were detected by chest computed tomographic scans in 28 patients (82%). Ground-glass opacities, which were a typical feature in adults, were rare in pediatric patients (3%). Rapid radiologic progression and a late-onset pattern of lesions in the lobules were also noticed. Lesions in lobules still existed in 24 (among 32 patients with lesions; 75%) patients that were discharged, although the main symptoms disappeared a few days after treatment. All patients were discharged, and the median duration of hospitalization was 10.00 (8.00-14.25) days. The current study was limited by the small sample size and a lack of dynamic detection of inflammatory markers. CONCLUSIONS: Our data systemically presented the clinical and epidemiological features, as well as the outcomes, of pediatric patients with COVID-19. Stratified analysis was performed between mild and moderate cases. The findings offer new insight into early identification and intervention in pediatric patients with COVID-19.


Coronavirus Infections/epidemiology , Coronavirus Infections/physiopathology , Lung/diagnostic imaging , Pneumonia, Viral/epidemiology , Pneumonia, Viral/physiopathology , Betacoronavirus , C-Reactive Protein/metabolism , COVID-19 , Child , Child, Preschool , China/epidemiology , Coinfection/epidemiology , Coronavirus Infections/diagnostic imaging , Coronavirus Infections/metabolism , Cough/epidemiology , Cough/physiopathology , Diarrhea/epidemiology , Diarrhea/physiopathology , Female , Fever/epidemiology , Fever/physiopathology , Humans , Hydroxybutyrate Dehydrogenase/metabolism , Infant , L-Lactate Dehydrogenase/metabolism , Length of Stay/statistics & numerical data , Male , Pandemics , Pneumonia, Viral/diagnostic imaging , Pneumonia, Viral/metabolism , Prealbumin/metabolism , Retrospective Studies , SARS-CoV-2 , Serum Amyloid A Protein/metabolism , Severity of Illness Index , Tomography, X-Ray Computed , Vomiting/epidemiology , Vomiting/physiopathology
19.
Mikrochim Acta ; 187(5): 277, 2020 04 20.
Article En | MEDLINE | ID: mdl-32314063

MXene nanosheets of type Ti3C2Tx were modified with ß-hydroxybutyrate dehydrogenase and then used as a biosensor for amperometric sensing of ß-hydroxybutyrate. The MXene and the nanocomposite were characterized by X-ray photoelectron spectroscopy, field-emission scanning electron microscopy, X-ray diffraction, and Fourier transform infrared spectroscopy. The MXene has a layered structure and proved to be an excellent immobilization matrix providing good compatibility with the enzyme ß-hydroxybutyrate dehydrogenase. The MXene-based biosensor, best operated at a potential of - 0.35 V (vs. Ag/AgCl), displays a wide linear range (0.36 to 17.9 mM), a sensitivity of 0.480 µA mM-1 cm-2, and a low detection limit (45 µM). The biosensor was successfully applied to the determination of ß-hydroxybutyrate in (spiked) real serum samples. Graphical abstract Schematic representation of the synthesis and decoration of Mxene 2D sheets with ß-hydroxybutyrate dehydrogenase for the amperometric determination of ß-hydroxybutyric acid.


3-Hydroxybutyric Acid/analysis , Carbon Compounds, Inorganic/chemistry , Hydroxybutyrate Dehydrogenase/chemistry , Nanocomposites/chemistry , Titanium/chemistry , 3-Hydroxybutyric Acid/metabolism , Biosensing Techniques , Electrochemical Techniques , Electrodes , Hydroxybutyrate Dehydrogenase/metabolism , Particle Size , Surface Properties
20.
J Inherit Metab Dis ; 43(5): 960-968, 2020 09.
Article En | MEDLINE | ID: mdl-32279332

d-3-Hydroxy-n-butyrate dehydrogenase (BDH1; EC 1.1.1.30), encoded by BDH1, catalyzes the reversible reduction of acetoacetate (AcAc) to 3-hydroxybutyrate (3HB). BDH1 is the last enzyme of hepatic ketogenesis and the first enzyme of ketolysis. The hereditary deficiency of BDH1 has not yet been described in humans. To define the features of BDH1 deficiency in a mammalian model, we generated Bdh1-deficient mice (Bdh1 KO mice). Under normal housing conditions, with unrestricted access to food, Bdh1 KO mice showed normal growth, appearance, behavior, and fertility. In contrast, fasting produced marked differences from controls. Although Bdh1 KO mice survive fasting for at least 48 hours, blood 3HB levels remained very low in Bdh1 KO mice, and despite AcAc levels moderately higher than in controls, total ketone body levels in Bdh1 KO mice were significantly lower than in wild-type (WT) mice after 16, 24, and 48 hours fasting. Hepatic fat content at 24 hours of fasting was greater in Bdh1 KO than in WT mice. Systemic BDH1 deficiency was well tolerated under normal fed conditions but manifested during fasting with a marked increase in AcAc/3HB ratio and hepatic steatosis, indicating the importance of ketogenesis for lipid energy balance in the liver.


Fasting/metabolism , Fatty Liver/genetics , Hydroxybutyrate Dehydrogenase/genetics , Ketone Bodies/metabolism , Liver/metabolism , Animals , Disease Models, Animal , Energy Metabolism , Fatty Liver/enzymology , Fatty Liver/physiopathology , Female , Hydroxybutyrate Dehydrogenase/deficiency , Hydroxybutyrate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
...