Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 83.132
Filtrer
1.
FASEB J ; 38(11): e23702, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38837439

RÉSUMÉ

Pyruvate kinase is a glycolytic enzyme that converts phosphoenolpyruvate and ADP into pyruvate and ATP. There are two genes that encode pyruvate kinase in vertebrates; Pkm and Pkl encode muscle- and liver/erythrocyte-specific forms, respectively. Each gene encodes two isoenzymes due to alternative splicing. Both muscle-specific enzymes, PKM1 and PKM2, function in glycolysis, but PKM2 also has been implicated in gene regulation due to its ability to phosphorylate histone 3 threonine 11 (H3T11) in cancer cells. Here, we examined the roles of PKM1 and PKM2 during myoblast differentiation. RNA-seq analysis revealed that PKM2 promotes the expression of Dpf2/Baf45d and Baf250a/Arid1A. DPF2 and BAF250a are subunits that identify a specific sub-family of the mammalian SWI/SNF (mSWI/SNF) of chromatin remodeling enzymes that is required for the activation of myogenic gene expression during differentiation. PKM2 also mediated the incorporation of DPF2 and BAF250a into the regulatory sequences controlling myogenic gene expression. PKM1 did not affect expression but was required for nuclear localization of DPF2. Additionally, PKM2 was required not only for the incorporation of phosphorylated H3T11 in myogenic promoters but also for the incorporation of phosphorylated H3T6 and H3T45 at myogenic promoters via regulation of AKT and protein kinase C isoforms that phosphorylate those amino acids. Our results identify multiple unique roles for PKM2 and a novel function for PKM1 in gene expression and chromatin regulation during myoblast differentiation.


Sujet(s)
Différenciation cellulaire , Histone , Myoblastes , Pyruvate kinase , Animaux , Pyruvate kinase/métabolisme , Pyruvate kinase/génétique , Souris , Phosphorylation , Histone/métabolisme , Histone/génétique , Myoblastes/métabolisme , Myoblastes/cytologie , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , , Humains , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/génétique , Hormones thyroïdiennes/métabolisme , Hormones thyroïdiennes/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Isoenzymes/métabolisme , Isoenzymes/génétique
3.
Carbohydr Res ; 541: 109169, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38838492

RÉSUMÉ

It is well established that tumour cells undergo metabolic changes to acquire biological advantage over normal cells with activation of the glycolytic pathway, a process termed "Warburg effect". Enzyme isoforms are alternative enzymatic forms with the same function but with different biochemical or epigenetic features. Moreover, isoforms may have varying impacts on different metabolic pathways. We challenge ourselves to analyse the glycolytic and gluconeogenic enzymes and isoforms in breast cancer, a complex and heterogeneous pathology, associated with high incidence and mortality rates especially among women. We analysed epithelial and tumour cell lines by RT-PCR and compared values to a publicly available database for the expression profile of normal and tumour tissues (Gepia) of enzymes and enzymatic isoforms from glycolytic and gluconeogenic pathways. Additionally, GeneMANIA was used to evaluate interactions, pathways, and attributes of each glycolytic/gluconeogenic steps. The findings reveal that the enzymes and enzymatic isoforms expressed in cell culture were somewhat different from those in breast tissue. We propose that the tumor microenvironment plays a crucial role in the expression of glycolytic and gluconeogenic enzymes and isoforms in tumour cells. Nonetheless, they not only participate in glycolytic and gluconeogenic enzymatic activities but may also influence other pathways, such as the Pentose-Phosphate-Pathway, TCA cycle, as well as other carbohydrate, lipid, and amino acid metabolism.


Sujet(s)
Tumeurs du sein , Néoglucogenèse , Glycolyse , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/enzymologie , Femelle , Lignée cellulaire tumorale , Isoenzymes/métabolisme , Isoenzymes/génétique
5.
Biol Pharm Bull ; 47(6): 1136-1143, 2024.
Article de Anglais | MEDLINE | ID: mdl-38866522

RÉSUMÉ

Ceramide (Cer) is synthesized de novo in the bilayer of the endoplasmic reticulum and transported to the cytosolic leaflet of the trans-Golgi apparatus for sphingomyelin (SM) synthesis. As the active site of SM synthase (SMS) is located on the luminal side of the Golgi membrane, Cer translocates to the lumen via transbilayer movement for SM synthesis. However, the mechanism of transbilayer movement is not fully understood. As the Cer-related translocases seem to localize near the SMS, the protein was identified using proximity-dependent biotin identification proteomics. Phospholipid scramblase 1 (PLSCR1), which is thought to act as a scramblase for phosphatidylserine and phosphatidylethanolamine, was identified as a protein proximal to the SMS isoforms SMS1 and SMS2. Although five isoforms of PLSCR have been reported in humans, only PLSCR1, PLSCR3, and PLSCR4 are expressed in HEK293T cells. Confocal microscopic analysis showed that PLSCR1 and PLSCR4 partially co-localized with p230, a trans-Golgi network marker, where SMS isoforms are localized. We established CRISPR/Cas9-mediated PLSCR1, PLSCR3, and PLSCR4 single-knockout cells and PLSCR1, 3, 4 triple knockout HEK293T cells. Liquid chromatography-tandem mass spectrometry revealed that the levels of species with distinct acyl chains in Cer and SM were not significantly different in single knockout cells or in the triple knockout cells compared to the wild-type cells. Our findings suggest that PLSCR1 is localized in the vicinity of SMS isoforms, however is not involved in the transbilayer movement of Cer for SM synthesis.


Sujet(s)
Protéines de transfert des phospholipides , Sphingomyéline , Transferases (other substituted phosphate groups) , Humains , Protéines de transfert des phospholipides/métabolisme , Protéines de transfert des phospholipides/génétique , Transferases (other substituted phosphate groups)/métabolisme , Transferases (other substituted phosphate groups)/génétique , Cellules HEK293 , Sphingomyéline/métabolisme , Sphingomyéline/biosynthèse , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Isoenzymes/métabolisme , Isoenzymes/génétique , Appareil de Golgi/métabolisme , Appareil de Golgi/enzymologie
6.
Free Radic Biol Med ; 220: 312-323, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38740101

RÉSUMÉ

Podocytes are crucial for regulating glomerular permeability. They have foot processes that are integral to the renal filtration barrier. Understanding their energy metabolism could shed light on the pathogenesis of filtration barrier injury. Lactate has been increasingly recognized as more than a waste product and has emerged as a significant metabolic fuel and reserve. The recent identification of lactate transporters in podocytes, the expression of which is modulated by glucose levels and lactate, highlights lactate's relevance. The present study investigated the impact of lactate on podocyte respiratory efficiency and mitochondrial dynamics. We confirmed lactate oxidation in podocytes, suggesting its role in cellular energy production. Under conditions of glucose deprivation or lactate supplementation, a significant shift was seen toward oxidative phosphorylation, reflected by an increase in the oxygen consumption rate/extracellular acidification rate ratio. Notably, lactate dehydrogenase A (LDHA) and lactate dehydrogenase B (LDHB) isoforms, which are involved in lactate conversion to pyruvate, were detected in podocytes for the first time. The presence of lactate led to higher intracellular pyruvate levels, greater LDH activity, and higher LDHB expression. Furthermore, lactate exposure increased mitochondrial DNA-to-nuclear DNA ratios and resulted in upregulation of the mitochondrial biogenesis markers peroxisome proliferator-activated receptor coactivator-1α and transcription factor A mitochondrial, regardless of glucose availability. Changes in mitochondrial size and shape were observed in lactate-exposed podocytes. These findings suggest that lactate is a pivotal energy source for podocytes, especially during energy fluctuations. Understanding lactate's role in podocyte metabolism could offer insights into renal function and pathologies that involve podocyte injury.


Sujet(s)
L-Lactate dehydrogenase , Acide lactique , Dynamique mitochondriale , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Podocytes , Podocytes/métabolisme , Podocytes/anatomopathologie , Animaux , Rats , Acide lactique/métabolisme , L-Lactate dehydrogenase/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Glucose/métabolisme , Métabolisme énergétique , Lactate dehydrogenase 5/métabolisme , Phosphorylation oxydative/effets des médicaments et des substances chimiques , ADN mitochondrial/métabolisme , ADN mitochondrial/génétique , Consommation d'oxygène , Cellules cultivées , Acide pyruvique/métabolisme , Isoenzymes
7.
Int J Mol Sci ; 25(10)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38791126

RÉSUMÉ

Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common metabolic disease of the liver, characterized by hepatic steatosis in more than 5% of hepatocytes. However, despite the recent approval of the first drug, resmetirom, for the management of metabolic dysfunction-associated steatohepatitis, decades of target exploration and hundreds of clinical trials have failed, highlighting the urgent need to find new druggable targets for the discovery of innovative drug candidates against MASLD. Here, we found that glutathione S-transferase alpha 1 (GSTA1) expression was negatively associated with lipid droplet accumulation in vitro and in vivo. Overexpression of GSTA1 significantly attenuated oleic acid-induced steatosis in hepatocytes or high-fat diet-induced steatosis in the mouse liver. The hepatoprotective and anti-inflammatory drug bicyclol also attenuated steatosis by upregulating GSTA1 expression. A detailed mechanism showed that GSTA1 directly interacts with fatty acid binding protein 1 (FABP1) and facilitates the degradation of FABP1, thereby inhibiting intracellular triglyceride synthesis by impeding the uptake and transportation of free fatty acids. Conclusion: GSTA1 may be a good target for the discovery of innovative drug candidates as GSTA1 stabilizers or enhancers against MASLD.


Sujet(s)
Protéines de liaison aux acides gras , Stéatose hépatique , Glutathione transferase , Régulation positive , Glutathione transferase/métabolisme , Glutathione transferase/génétique , Animaux , Humains , Souris , Protéines de liaison aux acides gras/métabolisme , Protéines de liaison aux acides gras/génétique , Stéatose hépatique/métabolisme , Stéatose hépatique/traitement médicamenteux , Régulation positive/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Mâle , Souris de lignée C57BL , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Métabolisme lipidique/effets des médicaments et des substances chimiques , Acide oléique/métabolisme , Cellules HepG2 , Triglycéride/métabolisme , Isoenzymes
8.
Int J Mol Sci ; 25(10)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38791142

RÉSUMÉ

Placenta Accreta Spectrum (PAS) is a life-threatening condition in which placental trophoblastic cells abnormally invade the uterus, often up to the uterine serosa and, in extreme cases, tissues beyond the uterine wall. Currently, there is no clinical assay for the non-invasive detection of PAS, and only ultrasound and MRI can be used for its diagnosis. Considering the subjectivity of visual assessment, the detection of PAS necessitates a high degree of expertise and, in some instances, can lead to its misdiagnosis. In clinical practice, up to 50% of pregnancies with PAS remain undiagnosed until delivery, and it is associated with increased risk of morbidity/mortality. Although many studies have evaluated the potential of fetal biomarkers circulating in maternal blood, very few studies have evaluated the potential of circulating placental extracellular vesicles (EVs) and their miRNA contents for molecular detection of PAS. Thus, to purify placental EVs from maternal blood, we customized our robust ultra-sensitive immuno-purification assay, termed EV-CATCHER, with a monoclonal antibody targeting the membrane Placental Alkaline Phosphatase (PLAP) protein, which is unique to the placenta and present on the surface of placental EVs. Then, as a pilot evaluation, we compared the miRNA expression profiles of placental EVs purified from the maternal plasma of women diagnosed with placenta previa (controls, n = 16); placenta lying low in uterus but not invasive) to those of placental EVs purified from the plasma of women with placenta percreta (cases, n = 16), PAS with the highest level of invasiveness. Our analyses reveal that miRNA profiling of PLAP+ EVs purified from maternal plasma identified 40 differentially expressed miRNAs when comparing these two placental pathologies. Preliminary miRNA pathway enrichment and gene ontology analysis of the top 14 upregulated and top nine downregulated miRNAs in PLAP+ EVs, purified from the plasma of women diagnosed with placenta percreta versus those diagnosed with placenta previa, suggests a potential role in control of cellular invasion and motility that will require further investigation.


Sujet(s)
Vésicules extracellulaires , Placenta accreta , Placenta , Humains , Femelle , Vésicules extracellulaires/métabolisme , Grossesse , Placenta/métabolisme , Placenta accreta/diagnostic , Placenta accreta/sang , Marqueurs biologiques/sang , Adulte , microARN/sang , microARN/génétique , microARN/métabolisme , Placenta previa/diagnostic , Placenta previa/sang , Phosphatase alcaline/métabolisme , Phosphatase alcaline/sang , Isoenzymes , Protéines liées au GPI
9.
Cell Signal ; 120: 111200, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38719019

RÉSUMÉ

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignancies. Lactate dehydrogenase family genes (LDHs) play a critical role in tumor metabolism, but their functions in HNSCC have not been investigated thoroughly. Thus, we aimed to explore the value of LDHs in HNSCC. METHODS: The association between LDHs expression and mutations, methylation, copy number variations (CNVs), alternative splicing (AS) and competing endogenous RNA (ceRNA) was investigated in The Cancer Genome Atlas (TCGA). The expression level of LDHs in OSCC tissues and adjacent normal tissues was verified by qPCR. Algorithms, such as ssGSEA, ESTIMATE, xCell and TIDE were utilized to analyze the characteristics of immune infiltration. Pathway alternations were enriched by GO, GSEA and KEGG analysis. The Mantel test was employed to elucidate the correlation between metabolism and the tumor microenvironment (TME). Subsequently, MTT and colony formation assays were utilized to assess the impact of LDHB knockdown on cellular proliferation. Additionally, ATP and lactate assays were performed to examine metabolic alterations. Co-culture experiments further investigated the effect of LDHB knockdown on T cell differentiation. RESULTS: LDHs were completely analyzed in multiple databases, among which LDHB was differentially expressed in HNSCC and significantly associated with prognosis. Low LDHB expression had better clinicopathological characteristics. Downregulated LDHB expression was associated with enhanced immune cell infiltration and could influence tumor metabolism. Despite having worse cytotoxic T lymphocyte dysfunction, the LDHBlow group was predicted to respond more favorably to immune checkpoint inhibitors (ICIs) therapy. Moreover, the correlation between metabolism and TME was depicted. In vitro, LDHB knockdown resulted in inhibited cell proliferation, increased lactate levels and decreased ATP levels, while promoted the Th1 differentiation of T cells. CONCLUSIONS: Our study provided a comprehensive analysis of the LDHs and illustrated low LDHB expression could inhibit tumor cell proliferation and ATP production by influencing metabolism, with improved immune cell infiltration and better response to immunotherapy.


Sujet(s)
Tumeurs de la tête et du cou , Immunothérapie , L-Lactate dehydrogenase , Carcinome épidermoïde de la tête et du cou , Humains , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , L-Lactate dehydrogenase/métabolisme , L-Lactate dehydrogenase/génétique , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/thérapie , Microenvironnement tumoral , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Différenciation cellulaire , Isoenzymes
10.
Protein J ; 43(3): 592-602, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38733555

RÉSUMÉ

The main structural difference between the mutation-susceptible retinal isoforms of inosine 5´-monophosphate dehydrogenase-1 (IMPDH-1) with the canonical form resides in the C- and N-terminal peptide extensions with unknown structural/functional impacts. In this report, we aimed to experimentally evaluate the functional impact of these extensions on the specific/non-specific single-stranded DNA (ssDNA)-binding activities relative to those of the canonical form. Our in silico findings indicated the possible contribution of the C-terminal segment to the reduced flexibility of the Bateman domain of the enzyme. In addition, the in silico data indicated that the N-terminal tail acts by altering the distance between the tetramers in the concave octamer complex (the native form) of the enzyme. The overall impact of these predicted structural variations became evident, first, through higher Km values with respect to either of the substrates relative to the canonical isoform, as reported previously (Andashti et al. in Mol Cell Biochem 465(1):155-164, 2020). Secondary, the binding of the recombinant mouse retinal isoform IMPDH1 (603) to its specific Rhodopsin target gene was significantly augmented while its binding to non-specific ssDNA was lower than that of the canonical isoform. The DNA-binding activity of the other mouse retinal isoform, IMPDH1(546), to specific and non-specific ssDNA was lower than that of the canonical form most probably due to the in silico predicted rigidity created in the Bateman domain by the C-terminal peptide extension. Furthermore, the DNA binding to the Rhodopsin target gene by each of the IMPDH isoforms influenced in the presence of GTP (Guanosine triphosphate) and ATP (Adenosine triphosphate).


Sujet(s)
IMP dehydrogenase , IMP dehydrogenase/métabolisme , IMP dehydrogenase/composition chimique , IMP dehydrogenase/génétique , Animaux , Souris , Isoenzymes/métabolisme , Isoenzymes/composition chimique , Isoenzymes/génétique , ADN simple brin/métabolisme , ADN simple brin/composition chimique , ADN simple brin/génétique , Rétine/métabolisme , Rétine/enzymologie , Liaison aux protéines , Humains
11.
Cancer Immunol Immunother ; 73(7): 127, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38739169

RÉSUMÉ

Lactate dehydrogenase B (LDHB) reversibly catalyzes the conversion of pyruvate to lactate or lactate to pyruvate and expressed in various malignancies. However, the role of LDHB in modulating immune responses against hepatocellular carcinoma (HCC) remains largely unknown. Here, we found that down-regulation of lactate dehydrogenase B (LDHB) was coupled with the promoter hypermethylation and knocking down the DNA methyltransferase 3A (DNMT 3A) restored LDHB expression levels in HCC cell lines. Bioinformatics analysis of the HCC cohort from The Cancer Genome Atlas revealed a significant positive correlation between LDHB expression and immune regulatory signaling pathways and immune cell infiltrations. Moreover, immune checkpoint inhibitors (ICIs) have shown considerable promise for HCC treatment and patients with higher LDHB expression responded better to ICIs. Finally, we found that overexpression of LDHB suppressed HCC growth in immunocompetent but not in immunodeficient mice, suggesting that the host immune system was involved in the LDHB-medicated tumor suppression. Our findings indicate that DNMT3A-mediated epigenetic silencing of LDHB may contribute to HCC progression through remodeling the tumor immune microenvironment, and LDHB may become a potential prognostic biomarker and therapeutic target for HCC immunotherapy.


Sujet(s)
Carcinome hépatocellulaire , DNA methyltransferase 3A , Épigenèse génétique , L-Lactate dehydrogenase , Tumeurs du foie , Microenvironnement tumoral , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Microenvironnement tumoral/immunologie , Humains , Animaux , Souris , L-Lactate dehydrogenase/métabolisme , L-Lactate dehydrogenase/génétique , DNA methyltransferase 3A/métabolisme , Régulation de l'expression des gènes tumoraux , Méthylation de l'ADN , Isoenzymes/génétique , Isoenzymes/métabolisme , Lignée cellulaire tumorale , Extinction de l'expression des gènes , Pronostic
12.
Ecotoxicol Environ Saf ; 279: 116500, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38795416

RÉSUMÉ

Hexavalent chromium [Cr(VI)] is one of the most common environmental contaminants due to its tremendous industrial applications, but its effects and mechanism remain to be investigated. Our previous studies showed that Cr(VI) exposure caused malignant transformation and tumorigenesis. This study showed that glycolytic proteins HK2 and LDHA levels were statistically significant changed in blood samples of Cr(VI)-exposed workers and in Cr-T cells compared to the control subjects and parental cells. HK2 and LDHA knockdown inhibited cell proliferation and angiogenesis, and higher HK2 and LDHA expression levels are associated with advanced stages and poor prognosis of lung cancer. We found that miR-218 levels were significantly decreased and miR-218 directly targeted HK2 and LDHA for inhibiting their expression. Overexpression of miR-218 inhibited glucose consumption and lactate production in Cr-T cells. Further study found that miR-218 inhibited tumor growth and angiogenesis by decreasing HK2 and LDHA expression in vivo. MiR-218 levels were negatively correlated with HK2 and LDHA expression levels and cancer development in human lung and other cancers. These results demonstrated that miR-218/HK2/LDHA pathway is vital for regulating Cr(VI)-induced carcinogenesis and human cancer development.


Sujet(s)
Carcinogenèse , Chrome , Hexokinase , Tumeurs du poumon , microARN , Régulation positive , microARN/génétique , Humains , Chrome/toxicité , Hexokinase/génétique , Hexokinase/métabolisme , Carcinogenèse/induit chimiquement , Tumeurs du poumon/induit chimiquement , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Pronostic , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , L-Lactate dehydrogenase/métabolisme , Exposition professionnelle/effets indésirables , Souris , Isoenzymes
13.
Acta Crystallogr D Struct Biol ; 80(Pt 6): 377-385, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38805243

RÉSUMÉ

Over the past forty years there has been a drastic increase in fructose-related diseases, including obesity, heart disease and diabetes. Ketohexokinase (KHK), the first enzyme in the liver fructolysis pathway, catalyzes the ATP-dependent phosphorylation of fructose to fructose 1-phosphate. Understanding the role of KHK in disease-related processes is crucial for the management and prevention of this growing epidemic. Molecular insight into the structure-function relationship in ligand binding and catalysis by KHK is needed for the design of therapeutic inhibitory ligands. Ketohexokinase has two isoforms: ketohexokinase A (KHK-A) is produced ubiquitously at low levels, whereas ketohexokinase C (KHK-C) is found at much higher levels, specifically in the liver, kidneys and intestines. Structures of the unliganded and liganded human isoforms KHK-A and KHK-C are known, as well as structures of unliganded and inhibitor-bound mouse KHK-C (mKHK-C), which shares 90% sequence identity with human KHK-C. Here, a high-resolution X-ray crystal structure of mKHK-C refined to 1.79 Šresolution is presented. The structure was determined in a complex with both the substrate fructose and the product of catalysis, ADP, providing a view of the Michaelis-like complex of the mouse ortholog. Comparison to unliganded structures suggests that KHK undergoes a conformational change upon binding of substrates that places the enzyme in a catalytically competent form in which the ß-sheet domain from one subunit rotates by 16.2°, acting as a lid for the opposing active site. Similar kinetic parameters were calculated for the mouse and human enzymes and indicate that mice may be a suitable animal model for the study of fructose-related diseases. Knowledge of the similarity between the mouse and human enzymes is important for understanding preclinical efforts towards targeting this enzyme, and this ground-state, Michaelis-like complex suggests that a conformational change plays a role in the catalytic function of KHK-C.


Sujet(s)
Fructokinases , Animaux , Fructokinases/composition chimique , Fructokinases/métabolisme , Souris , Cristallographie aux rayons X , Isoenzymes/composition chimique , Modèles moléculaires , Conformation des protéines , Humains , Fructose/métabolisme , Fructose/composition chimique
14.
J Transl Med ; 22(1): 474, 2024 May 19.
Article de Anglais | MEDLINE | ID: mdl-38764020

RÉSUMÉ

BACKGROUND: The initiation of fibroblast growth factor 1 (FGF1) expression coincident with the decrease of FGF2 expression is a well-documented event in prostate cancer (PCa) progression. Lactate dehydrogenase A (LDHA) and LDHB are essential metabolic products that promote tumor growth. However, the relationship between FGF1/FGF2 and LDHA/B-mediated glycolysis in PCa progression is not reported. Thus, we aimed to explore whether FGF1/2 could regulate LDHA and LDHB to promote glycolysis and explored the involved signaling pathway in PCa progression. METHODS: In vitro studies used RT‒qPCR, Western blot, CCK-8 assays, and flow cytometry to analyze gene and protein expression, cell viability, apoptosis, and cell cycle in PCa cell lines. Glycolysis was assessed by measuring glucose consumption, lactate production, and extracellular acidification rate (ECAR). For in vivo studies, a xenograft mouse model of PCa was established and treated with an FGF pathway inhibitor, and tumor growth was monitored. RESULTS: FGF1, FGF2, and LDHA were expressed at high levels in PCa cells, while LDHB expression was low. FGF1/2 positively modulated LDHA and negatively modulated LDHB in PCa cells. The depletion of FGF1, FGF2, or LDHA reduced cell proliferation, induced cell cycle arrest, and inhibited glycolysis. LDHB overexpression showed similar inhibitory effect on PCa cells. Mechanistically, we found that FGF1/2 positively regulated STAT1 and STAT1 transcriptionally activated LDHA expression while suppressed LDHB expression. Furthermore, the treatment of an FGF pathway inhibitor suppressed PCa tumor growth in mice. CONCLUSION: The FGF pathway facilitates glycolysis by activating LDHA and suppressing LDHB in a STAT1-dependent manner in PCa.


Sujet(s)
Facteurs de croissance fibroblastique , Glycolyse , L-Lactate dehydrogenase , Tumeurs de la prostate , Facteur de transcription STAT-1 , Transduction du signal , Mâle , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Humains , Animaux , L-Lactate dehydrogenase/métabolisme , Lignée cellulaire tumorale , Facteur de transcription STAT-1/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Souris nude , Prolifération cellulaire , Souris , Régulation de l'expression des gènes tumoraux , Facteur de croissance fibroblastique de type 2/métabolisme , Apoptose , Lactate dehydrogenase 5/métabolisme , Isoenzymes
15.
CNS Neurosci Ther ; 30(5): e14741, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38702940

RÉSUMÉ

AIMS: Despite the success of single-cell RNA sequencing in identifying cellular heterogeneity in ischemic stroke, clarifying the mechanisms underlying these associations of differently expressed genes remains challenging. Several studies that integrate gene expression and gene expression quantitative trait loci (eQTLs) with genome wide-association study (GWAS) data to determine their causal role have been proposed. METHODS: Here, we combined Mendelian randomization (MR) framework and single cell (sc) RNA sequencing to study how differently expressed genes (DEGs) mediating the effect of gene expression on ischemic stroke. The hub gene was further validated in the in vitro model. RESULTS: We identified 2339 DEGs in 10 cell clusters. Among these DEGs, 58 genes were associated with the risk of ischemic stroke. After external validation with eQTL dataset, lactate dehydrogenase B (LDHB) is identified to be positively associated with ischemic stroke. The expression of LDHB has also been validated in sc RNA-seq with dominant expression in microglia and astrocytes, and melatonin is able to reduce the LDHB expression and activity in vitro ischemic models. CONCLUSION: Our study identifies LDHB as a novel biomarker for ischemic stroke via combining the sc RNA-seq and MR analysis.


Sujet(s)
Accident vasculaire cérébral ischémique , L-Lactate dehydrogenase , Mélatonine , Analyse de randomisation mendélienne , Analyse de séquence d'ARN , Animaux , Humains , Étude d'association pangénomique/méthodes , Accident vasculaire cérébral ischémique/génétique , Accident vasculaire cérébral ischémique/métabolisme , Isoenzymes/génétique , Isoenzymes/métabolisme , L-Lactate dehydrogenase/métabolisme , L-Lactate dehydrogenase/génétique , Analyse de randomisation mendélienne/méthodes , Locus de caractère quantitatif , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Souris
16.
Chemosphere ; 358: 142249, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38705405

RÉSUMÉ

Chlorophenols (CPs) are a group of pollutants that pose a great threat to the environment, they are widely used in industrial and agricultural wastes, pesticides, herbicides, textiles, pharmaceuticals and plastics. Among CPs, pentachlorophenol was listed as one of the persistent organic pollutants (POPs) by the Stockholm convention. This study aims to identify the UDP-glucosyltransferase (UGT) isoforms involved in the metabolic elimination of CPs. CPs' mono-glucuronide was detected in the human liver microsomes (HLMs) incubation mixture with co-factor uridine-diphosphate glucuronic acid (UDPGA). HLMs-catalyzed glucuronidation metabolism reaction equations followed Michaelis-Menten or substrate inhibition type. Recombinant enzymes and chemical reagents inhibition experiments were utilized to phenotype the main UGT isoforms involved in the glucuronidation of CPs. UGT1A6 might be the major enzyme in the glucuronidation of mono-chlorophenol isomer. UGT1A1, UGT1A6, UGT1A9, UGT2B4 and UGT2B7 were the most important five UGT isoforms for metabolizing the di-chlorophenol and tri-chlorophenol isomers. UGT1A1 and UGT1A3 were the most important UGT isoforms in the catalysis of tetra-chlorophenol and pentachlorophenol isomers. Species differences were investigated using rat liver microsomes (RLMs), pig liver microsomes (PLMs), dog liver microsomes (DLMs), and monkey liver microsomes (MyLMs). All these results were helpful for elucidating the metabolic elimination and toxicity of CPs.


Sujet(s)
Chlorophénols , Glucuronosyltransferase , Microsomes du foie , Glucuronosyltransferase/métabolisme , Chlorophénols/métabolisme , Animaux , Microsomes du foie/métabolisme , Humains , Rats , Polluants environnementaux/métabolisme , Isoenzymes/métabolisme , Glucuronides/métabolisme
17.
Cell Rep ; 43(4): 114047, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38607916

RÉSUMÉ

Using 13C6 glucose labeling coupled to gas chromatography-mass spectrometry and 2D 1H-13C heteronuclear single quantum coherence NMR spectroscopy, we have obtained a comparative high-resolution map of glucose fate underpinning ß cell function. In both mouse and human islets, the contribution of glucose to the tricarboxylic acid (TCA) cycle is similar. Pyruvate fueling of the TCA cycle is primarily mediated by the activity of pyruvate dehydrogenase, with lower flux through pyruvate carboxylase. While the conversion of pyruvate to lactate by lactate dehydrogenase (LDH) can be detected in islets of both species, lactate accumulation is 6-fold higher in human islets. Human islets express LDH, with low-moderate LDHA expression and ß cell-specific LDHB expression. LDHB inhibition amplifies LDHA-dependent lactate generation in mouse and human ß cells and increases basal insulin release. Lastly, cis-instrument Mendelian randomization shows that low LDHB expression levels correlate with elevated fasting insulin in humans. Thus, LDHB limits lactate generation in ß cells to maintain appropriate insulin release.


Sujet(s)
Sécrétion d'insuline , Cellules à insuline , L-Lactate dehydrogenase , Acide lactique , Humains , Cellules à insuline/métabolisme , Animaux , L-Lactate dehydrogenase/métabolisme , Souris , Acide lactique/métabolisme , Glucose/métabolisme , Insuline/métabolisme , Isoenzymes/métabolisme , Cycle citrique , Souris de lignée C57BL , Mâle
19.
Pflugers Arch ; 476(7): 1041-1064, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38658400

RÉSUMÉ

Signaling of G protein-activated inwardly rectifying K+ (GIRK) channels is an important mechanism of the parasympathetic regulation of the heart rate and cardiac excitability. GIRK channels are inhibited during stimulation of Gq-coupled receptors (GqPCRs) by depletion of phosphatidyl-4,5-bisphosphate (PIP2) and/or channel phosphorylation by protein kinase C (PKC). The GqPCR-dependent modulation of GIRK currents in terms of specific PKC isoform activation was analyzed in voltage-clamp experiments in rat atrial myocytes and in CHO or HEK 293 cells. By using specific PKC inhibitors, we identified the receptor-activated PKC isoforms that contribute to phenylephrine- and angiotensin-induced GIRK channel inhibition. We demonstrate that the cPKC isoform PKCα significantly contributes to GIRK inhibition during stimulation of wildtype α1B-adrenergic receptors (α1B-ARs). Deletion of the α1B-AR serine residues S396 and S400 results in a preferential regulation of GIRK activity by PKCß. As a novel finding, we report that the AT1-receptor-induced GIRK inhibition depends on the activation of the nPKC isoform PKCε whereas PKCα and PKCß do not mainly participate in the angiotensin-mediated GIRK reduction. Expression of the dominant negative (DN) PKCε prolonged the onset of GIRK inhibition and significantly reduced AT1-R desensitization, indicating that PKCε regulates both GIRK channel activity and the strength of the receptor signal via a negative feedback mechanism. The serine residue S418 represents an important phosphorylation site for PKCε in the GIRK4 subunit. To analyze the functional impact of this PKC phosphorylation site for receptor-specific GIRK channel modulation, we monitored the activity of a phosphorylation-deficient (GIRK4 (S418A)) GIRK4 channel mutant during stimulation of α1B-ARs or AT1-receptors. Mutation of S418 did not impede α1B-AR-mediated GIRK inhibition, suggesting that S418 within the GIRK4 subunit is not subject to PKCα-induced phosphorylation. Furthermore, activation of angiotensin receptors induced pronounced GIRK4 (S418A) channel inhibition, excluding that this phosphorylation site contributes to the AT1-R-induced GIRK reduction. Instead, phosphorylation of S418 has a facilitative effect on GIRK activity that was abolished in the GIRK4 (S418A) mutant. To summarize, the present study shows that the receptor-dependent regulation of atrial GIRK channels is attributed to the GqPCR-specific activation of different PKC isoforms. Receptor-specific activated PKC isoforms target distinct phosphorylation sites within the GIRK4 subunit, resulting in differential regulation of GIRK channel activity with either facilitative or inhibitory effects on GIRK currents.


Sujet(s)
Cricetulus , Canaux potassiques rectifiants entrants couplés aux protéines G , Protéine kinase C , Canaux potassiques rectifiants entrants couplés aux protéines G/métabolisme , Animaux , Phosphorylation , Cellules HEK293 , Humains , Rats , Protéine kinase C/métabolisme , Cellules CHO , Récepteurs alpha-1 adrénergiques/métabolisme , Myocytes cardiaques/métabolisme , Mâle , Rat Wistar , Protein kinase C-alpha/métabolisme , Isoenzymes/métabolisme
20.
Cancer Lett ; 590: 216869, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38593918

RÉSUMÉ

Lysine acetyltransferase 7 (KAT7), a histone acetyltransferase, has recently been identified as an oncoprotein and has been implicated in the development of various malignancies. However, its specific role in head and neck squamous carcinoma (HNSCC) has not been fully elucidated. Our study revealed that high expression of KAT7 in HNSCC patients is associated with poor survival prognosis and silencing KAT7 inhibits the Warburg effect, leading to reduced proliferation, invasion, and metastatic potential of HNSCC. Further investigation uncovered a link between the high expression of KAT7 in HNSCC and tumor-specific glycolytic metabolism. Notably, KAT7 positively regulates Lactate dehydrogenase A (LDHA), a key enzyme in metabolism, to promote lactate production and create a conducive environment for tumor proliferation and metastasis. Additionally, KAT7 enhances LDHA activity and upregulates LDHA protein expression by acetylating the lysine 118 site of LDHA. Treatment with WM3835, a KAT7 inhibitor, effectively suppressed the growth of subcutaneously implanted HNSCC cells in mice. In conclusion, our findings suggest that KAT7 exerts pro-cancer effects in HNSCC by acetylating LDHA and may serve as a potential therapeutic target. Inhibiting KAT7 or LDHA expression holds promise as a therapeutic strategy to suppress the growth and progression of HNSCC.


Sujet(s)
Prolifération cellulaire , Tumeurs de la tête et du cou , Histone acetyltransferases , Carcinome épidermoïde de la tête et du cou , Humains , Animaux , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/métabolisme , Acétylation , Lignée cellulaire tumorale , Histone acetyltransferases/métabolisme , Histone acetyltransferases/génétique , Souris , L-Lactate dehydrogenase/métabolisme , L-Lactate dehydrogenase/génétique , Lysine acetyltransferases/métabolisme , Lysine acetyltransferases/génétique , Régulation de l'expression des gènes tumoraux , Souris nude , Effet Warburg en oncologie , Mâle , Femelle , Mouvement cellulaire , Tests d'activité antitumorale sur modèle de xénogreffe , Invasion tumorale , Isoenzymes/métabolisme , Isoenzymes/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...