Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 101
Filtrer
1.
Adv Nutr ; 15(4): 100200, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38438107

RÉSUMÉ

The epithelium lining the intestinal tract serves a multifaceted role. It plays a crucial role in nutrient absorption and immune regulation and also acts as a protective barrier, separating underlying tissues from the gut lumen content. Disruptions in the delicate balance of the gut epithelium trigger inflammatory responses, aggravate conditions such as inflammatory bowel disease, and potentially lead to more severe complications such as colorectal cancer. Maintaining intestinal epithelial homeostasis is vital for overall health, and there is growing interest in identifying nutraceuticals that can strengthen the intestinal epithelium. α-Ketoglutarate, a metabolite of the tricarboxylic acid cycle, displays a variety of bioactive effects, including functioning as an antioxidant, a necessary cofactor for epigenetic modification, and exerting anti-inflammatory effects. This article presents a comprehensive overview of studies investigating the potential of α-ketoglutarate supplementation in preventing dysfunction of the intestinal epithelium.


Sujet(s)
Maladies inflammatoires intestinales , Acides cétoglutariques , Humains , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/usage thérapeutique , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/prévention et contrôle , Muqueuse intestinale
2.
Mol Med ; 30(1): 15, 2024 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-38254035

RÉSUMÉ

BACKGROUND: In heart failure (HF), mitochondrial dysfunction and metabolic remodeling lead to a reduction in energy productivity and aggravate cardiomyocyte injury. Supplementation with α-ketoglutarate (AKG) alleviated myocardial hypertrophy and fibrosis in mice with HF and improved cardiac insufficiency. However, the myocardial protective mechanism of AKG remains unclear. We verified the hypothesis that AKG improves mitochondrial function by upregulating NAD+ levels and activating silent information regulator 2 homolog 1 (SIRT1) in cardiomyocytes. METHODS: In vivo, 2% AKG was added to the drinking water of mice undergoing transverse aortic constriction (TAC) surgery. Echocardiography and biopsy were performed to evaluate cardiac function and pathological changes. Myocardial metabolomics was analyzed by liquid chromatography‒mass spectrometry (LC‒MS/MS) at 8 weeks after surgery. In vitro, the expression of SIRT1 or PINK1 proteins was inhibited by selective inhibitors and siRNA in cardiomyocytes stimulated with angiotensin II (AngII) and AKG. NAD+ levels were detected using an NAD test kit. Mitophagy and ferroptosis levels were evaluated by Western blotting, qPCR, JC-1 staining and lipid peroxidation analysis. RESULTS: AKG supplementation after TAC surgery could alleviate myocardial hypertrophy and fibrosis and improve cardiac function in mice. Metabolites of the malate-aspartate shuttle (MAS) were increased, but the TCA cycle and fatty acid metabolism pathway could be inhibited in the myocardium of TAC mice after AKG supplementation. Decreased NAD+ levels and SIRT1 protein expression were observed in heart of mice and AngII-treated cardiomyocytes. After AKG treatment, these changes were reversed, and increased mitophagy, inhibited ferroptosis, and alleviated damage in cardiomyocytes were observed. When the expression of SIRT1 was inhibited by a selective inhibitor and siRNA, the protective effect of AKG was suppressed. CONCLUSION: Supplementation with AKG can improve myocardial hypertrophy, fibrosis and chronic cardiac insufficiency caused by pressure overload. By increasing the level of NAD+, the SIRT-PINK1 and SIRT1-GPX4 signaling pathways are activated to promote mitophagy and inhibit ferroptosis in cardiomyocytes, which ultimately alleviates cardiomyocyte damage.


Sujet(s)
Sténose aortique , Ferroptose , Défaillance cardiaque , Acides cétoglutariques , Mitophagie , Angiotensine-II , Chromatographie en phase liquide , Ferroptose/effets des médicaments et des substances chimiques , Fibrose , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Hypertrophie , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/usage thérapeutique , Mitophagie/effets des médicaments et des substances chimiques , Myocytes cardiaques , NAD , Protein kinases , Petit ARN interférent , Sirtuine-1 , Spectrométrie de masse en tandem , Animaux , Souris
3.
Acta Biomater ; 173: 442-456, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-37984632

RÉSUMÉ

Osteoporosis (OP), which largely increases the risk of fractures, is the most common chronic degenerative orthopedic disease in the elderly due to the imbalance of bone homeostasis. Alpha-ketoglutaric acid (AKG), an endogenous metabolic intermediate involved in osteogenesis, plays critical roles in osteogenic differentiation and mineralization and the inhibition of osteoclastogenic differentiation. However, the low bioavailability and poor bone-targeting efficiency of AKG seriously limit its efficacy in OP treatment. In this work, a bone-targeting, near-infrared emissive lanthanide luminescence nanocarrier loaded with AKG (ß-NaYF4:7%Yb, 60%Nd@NaLuF4@mSiO2-EDTA-AKG, abbreviated as LMEK) is developed for the enhancement of AKG efficacy in OP therapy. By utilizing the NIR-II luminescence (>1000 nm) of LMEK, whole-body bone imaging with high spatial resolution is achieved to confirm the bone enrichment of AKG noninvasively in vivo. The results reveal that LMEK exhibits a remarkable OP therapeutic effect in improving the osseointegration of the surrounding bone in the ovariectomized OP mice models, which is validated by the enhanced inhibition of osteoclast through hypoxia-inducible factor-1α suppression and promotion of osteogenic differentiation in osteoblast. Notably, the dose of AKG in LMEK can be reduced to only 0.2 % of the dose when pure AKG is used in therapy, which dramatically improves the bioavailability of AKG and mitigates the metabolism burden. This work provides a strategy to conquer the low utilization of AKG in OP therapy, which not only overcomes the challenges in AKG efficacy for OP treatment but also offers insights into the development and application of other potential drugs for skeletal diseases. STATEMENT OF SIGNIFICANCE: Alpha-ketoglutarate (AKG) is an intermediate within the Krebs cycle, participating in diverse metabolic and cellular processes, showing potential for osteoporosis (OP) therapy. However, AKG's limited bioavailability and inefficient bone-targeting hinder its effectiveness in treating OP. Herein, a near-infrared emissive nanocarrier is developed that precisely targets bones and delivers AKG, bolstering its effectiveness in OP therapy. Thanks to this efficient bone-targeting delivery, the AKG dosage is reduced to 0.2 % of the conventional treatment level. This marks the first utilization of a bone-targeting nanocarrier to amplify AKG's bioavailability and OP therapy efficacy. Furthermore, the mechanism of AKG-loaded nanocarrier regulating the biological behavior of osteoclasts and osteoblasts mediated is tentatively explored.


Sujet(s)
Acides cétoglutariques , Ostéoporose , Humains , Souris , Animaux , Sujet âgé , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/métabolisme , Acides cétoglutariques/usage thérapeutique , Ostéogenèse , Luminescence , Ostéoporose/traitement médicamenteux , Ostéoblastes/métabolisme
4.
Eur J Pharmacol ; 960: 176118, 2023 Dec 05.
Article de Anglais | MEDLINE | ID: mdl-37871764

RÉSUMÉ

Gastric lesions have several aetiologies, among which stress is the most prominent. Therefore, identification of new therapies to prevent stress is of considerable importance. Alpha-ketoglutarate (α-kg) several beneficial effects and has shown promise in combating oxidative stress, inflammation, and premature aging. Thus, this study aimed to evaluate the protective effect of α-kg in a gastric damage model by water-immersion restraint stress (WIRS). Pretreatment with α-kg decreased stress-related histopathological scores of tissue oedema, cell loss, and inflammatory infiltration. The α-kg restored the percentage of type III collagen fibres. Mucin levels were preserved as well as the structure and area of the myenteric plexus ganglia were preserved after pretreatment with α-kg. Myeloperoxidase (MPO) levels and the expression of pro-inflammatory cytokines (TNF-α and IL-1ß) were also reduced following α-kg pretreatment. Decreased levels of glutathione (GSH) in the stress group were restored by α-kg. The omeprazole group was used as standard drug e also demonstrated improve on some parameters after the exposition to WIRS as inflammatory indexes, GSH and mucin. Through this, was possible to observe that α-kg can protect the gastric mucosa exposed to WIRS, preserve tissue architecture, reduce direct damage to the mucosa and inflammatory factors, stimulate the production of type III collagen and mucin, preserve the myenteric plexus ganglia, and maintain antioxidant potential. Due to, we indicate that α-kg has protective activity of the gastric mucosa, demonstrating its ability to prevent damage associated with gastric lesions caused by stress.


Sujet(s)
Acides cétoglutariques , Ulcère gastrique , Souris , Animaux , Acides cétoglutariques/métabolisme , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/usage thérapeutique , Ulcère gastrique/anatomopathologie , Collagène de type III/métabolisme , Immersion , Muqueuse gastrique , Glutathion/métabolisme , Mucines/métabolisme , Eau/métabolisme , Contention physique/effets indésirables
5.
Redox Biol ; 62: 102663, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36924682

RÉSUMÉ

Osteoarthritis (OA) is an age-related metabolic disease. Low-grade inflammation and oxidative stress are the last common pathway of OA. α-ketoglutarate (α-KG) is an essential physiological metabolite from the mitochondrial tricarboxylic acid (TCA) cycle, with multiple functions, including anti-inflammation and antioxidation, and exhibits decreased serum levels with age. Herein, we aimed to investigate the effect and mechanism of α-KG on OA. We first quantified the α-KG levels in human cartilage tissue and osteoarthritic chondrocytes induced by IL-1ß. Besides, IL-1ß-induced osteoarthritic chondrocytes were treated with different concentrations of α-KG. Chondrocyte proliferation and apoptosis, synthesis and degradation of extracellular matrix, and inflammation mediators were analyzed. RNA sequencing was used to explore the mechanism of α-KG, and mitophagy and oxidative stress levels were further detected. These results were verified in an anterior cruciate ligament transection (ACLT) induced age-related OA rat model. We found that α-KG content decreased by 31.32% in damaged medial cartilage than in normal lateral cartilage and by 36.85% in IL-1ß-induced human osteoarthritic chondrocytes compared to control. α-KG supplementation reversed IL-1ß-induced chondrocyte proliferation inhibition and apoptosis, increased the transcriptomic and proteinic expression of ACAN and COL2A1 in vivo and in vitro, but inhibited the expression of MMP13, ADAMTS5, IL-6, and TNF-α. In mechanism, α-KG promoted mitophagy and inhibited ROS generation, and these effects could be prevented by Mdivi-1 (a mitophagy inhibitor). Overall, α-KG content decreased in human OA cartilage and IL-1ß-induced osteoarthritic chondrocytes. Moreover, α-KG supplementation could alleviate osteoarthritic phenotype by regulating mitophagy and oxidative stress, suggesting its potential as a therapeutic target to ameliorate OA.


Sujet(s)
Acides cétoglutariques , Arthrose , Humains , Rats , Animaux , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/métabolisme , Acides cétoglutariques/usage thérapeutique , Mitophagie , Arthrose/traitement médicamenteux , Arthrose/métabolisme , Chondrocytes/métabolisme , Stress oxydatif , Interleukine-1 bêta/métabolisme , Cellules cultivées
6.
Cell Death Dis ; 14(2): 170, 2023 02 28.
Article de Anglais | MEDLINE | ID: mdl-36854755

RÉSUMÉ

Patients with advanced melanoma have shown an improved outlook after anti-PD1 therapy, but the low response rate restricts clinical benefit; therefore, enhancing anti-PD1 therapeutic efficacy remains a major challenge. Here, our findings showed a significantly increased abundance of α-KG in healthy controls, anti-PD1-sensitive melanoma-bearing mice, and anti-PD1-sensitive melanoma patients; moreover, supplementation with α-KG enhanced the efficacy of anti-PD1 immunotherapy and increased PD-L1 expression in melanoma tumors via STAT1/3. We also found that supplementation with α-KG significantly increased the activity of the methylcytosine dioxygenases TET2/3, which led to an increased 5-hydroxymethylcytosine (5-hmC) level in the PD-L1 promoter. As a consequence, STAT1/3 binding to the PD-L1 promoter was stabilized to upregulate PD-L1 expression. Importantly, single-cell sequencing of preclinical samples and analysis of clinical data revealed that TET2/3-STAT1/3-CD274 signaling was associated with sensitivity to anti-PD1 treatment in melanoma. Taken together, our results provide novel insight into α-KG's function in anti-PD1 treatment of melanoma and suggest supplementation with α-KG as a novel promising strategy to improve the efficacy of anti-PD1 therapy.


Sujet(s)
Antigène CD274 , Acides cétoglutariques , Mélanome , Animaux , Souris , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/génétique , Compléments alimentaires , Épigenèse génétique , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/usage thérapeutique , Mélanome/traitement médicamenteux , Mélanome/génétique
7.
Environ Toxicol ; 38(1): 17-27, 2023 Jan.
Article de Anglais | MEDLINE | ID: mdl-36111830

RÉSUMÉ

BACKGROUND: Alpha-ketoglutarate (AKG) or 2-oxoglutarate is a key substance in the tricarboxylic acid cycle (TCA) and has been known to play an important role in cancerogenesis and tumor progression. Renal cell carcinoma (RCC) is the most common type of kidney cancer, and it has a high mortality rate. Autophagy is a phenomenon of self-digestion, and its significance in tumor genesis and progression remains debatable. However, the mechanisms underlying how AKG regulates autophagy in RCC remain unknown. Thus, the purpose of this study was to assess the therapeutic efficacy of AKG and its molecular mechanisms. METHODS: RCC cell lines 786O and ACHN were treated with varying doses of AKG for 24 h. CCK-8, Transwell, and scratch wound healing assays were utilized to evaluate the role of AKG in RCC cells. Autophagy protein and PI3K/AKT/mTOR pathway protein levels were analyzed by Western blot. RESULTS: AKG inhibited the proliferation of RCC cells 786O and ACHN in a dose-dependent manner according to the CCK-8 assay. In addition, flow cytometry and Western blot analysis revealed that AKG dose-dependently triggered apoptosis and autophagy in RCC cells. By promoting cell apoptosis and autophagy, AKG dramatically suppressed tumor growth. Mechanistically, AKG induces autophagy by promoting ROS generation and inhibiting the PI3K/AKT/mTOR pathway. CONCLUSIONS: The anti-tumor effect of AKG promotes autophagy in renal cancer cells via mediating ROS-PI3K/Akt/mTOR, and may be used as a potential anticancer drug for kidney cancer.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Humains , Néphrocarcinome/anatomopathologie , Espèces réactives de l'oxygène , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/usage thérapeutique , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal , Prolifération cellulaire , Sérine-thréonine kinases TOR/métabolisme , Autophagie , Apoptose , Stress du réticulum endoplasmique , Tumeurs du rein/anatomopathologie
8.
Biomater Sci ; 10(23): 6688-6697, 2022 Nov 22.
Article de Anglais | MEDLINE | ID: mdl-36190458

RÉSUMÉ

Activated effector T cells induce pro-inflammatory responses in rheumatoid arthritis (RA) which then lead to inflammation of the joints. In this report, we demonstrate that polymeric nanoparticles with alpha keto-glutarate (aKG) in their polymer backbone (termed as paKG NPs) modulate T cell responses in vitro and in vivo. Impressively, a low dose of only three administrations of methotrexate, a clinically and chronically administered drug for RA, in conjunction with two doses of paKG NPs, reversed arthritis symptoms in collagen-induced arthritis (CIA) mice. This was further followed by significant decreases in pro-inflammatory antigen-specific T helper type 17 (TH17) responses and a significant increase in anti-inflammatory regulatory T cell (TREG) responses when CIA treated splenic cells were isolated and re-exposed to the CIA self-antigen. Overall, this study supports the concurrent and short term, low dose of paKG NPs and methotrexate for the reversal of RA symptoms.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Nanoparticules , Souris , Animaux , Méthotrexate/pharmacologie , Méthotrexate/usage thérapeutique , Acides cétoglutariques/usage thérapeutique , Souris de lignée DBA , Polyarthrite rhumatoïde/traitement médicamenteux , Arthrite expérimentale/induit chimiquement , Arthrite expérimentale/traitement médicamenteux , Lymphocytes T auxiliaires/métabolisme , Polymères/usage thérapeutique
9.
Int Immunopharmacol ; 112: 109190, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36116152

RÉSUMÉ

Macrophages exhibited different phenotypes in response to environmental cues. To meet the needs of rapid response to stimuli, M1-activated macrophages preferred glycolysis to oxidative phosphorylation (OXPHOS) in mitochondria to quickly produce energy and obtain ample raw materials to support cell activation at the same time. Activated macrophages produced free radicals and cytokines to eradicate pathogens but also induced oxidative damage and enhanced inflammation. Grossamide (GSE), a lignanamide from Polygonum multiflorum Thunb., exhibited notable anti-inflammatory effects. In this study, the potential of GSE on macrophage polarization was explored. GSE significantly down-regulated the levels of M1 macrophage biomarkers (Cd32a, Cd80 and Cd86) while increased the levels of M2 indicators (Cd163, Mrc1 and Socs1), showing its potential to inhibit LPS-induced M1 polarization of macrophages. This ability has close a link to its effect on metabolic reprogramming of macrophage. GSE shunted nitric oxide (NO) production from arginine by up-regulation of arginase and down-regulation of inducible nitric oxide synthase, thus attenuated the inhibition of NO on OXPHOS. LPS created three breakpoints in the tricarboxylic acid cycle (TCA) cycle of macrophage as evidenced by down-regulated isocitrate dehydrogenase, accumulation of succinate and the inhibited SDH activity, significantly decreased level of oxoglutarate dehydrogenase expression and its substrate α-ketoglutarate. Thus GSE reduced oxidative stress and amended fragmented TCA cycle. As a result, GSE maintained redox (NAD+/NADH) and energy (ATP/ADP) state, reduced extracellular acidification rate and enhanced the oxygen consumption rate. In addition, GSE decreased the release of inflammatory cytokines by inhibiting the activation of the LPS/TLR4/NF-κB pathway. These findings highlighted the central role of immunometabolism of macrophages in its functional plasticity, which invited future study of mode of action of anti-inflammatory drugs from viewpoint of metabolic reprogramming.


Sujet(s)
NAD , Facteur de transcription NF-kappa B , Souris , Animaux , Nitric oxide synthase type II/métabolisme , NAD/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Lipopolysaccharides/pharmacologie , Monoxyde d'azote/métabolisme , Arginase/métabolisme , Récepteur de type Toll-4/métabolisme , Isocitrate dehydrogenases/métabolisme , Isocitrate dehydrogenases/pharmacologie , Isocitrate dehydrogenases/usage thérapeutique , Acides cétoglutariques/métabolisme , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/usage thérapeutique , Activation des macrophages , Macrophages , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Cytokines/métabolisme , Anti-inflammatoires/usage thérapeutique , Succinates/usage thérapeutique , Ketoglutarate dehydrogenase complex/métabolisme , Ketoglutarate dehydrogenase complex/pharmacologie , Arginine/usage thérapeutique , ADP/métabolisme , ADP/pharmacologie , ADP/usage thérapeutique , Adénosine triphosphate/métabolisme
10.
Exp Gerontol ; 168: 111944, 2022 10 15.
Article de Anglais | MEDLINE | ID: mdl-36064157

RÉSUMÉ

BACKGROUND: Metabolic alteration is a mainstream concept underlying the cognitive decline in neurodegenerative disorders including Alzheimer's disease (AD). Mitochondrial enzyme α-ketoglutarate dehydrogenase complex (α-KGDHC) seems to play a dual-edged sword role in cytotoxic insult. Here, using succinyl phosphonate (SP), a specific α-KGDHC inhibitor, we aimed to examine its potential action on AD progression. METHODS: Male Wistar rats were assigned to two separate experiments. First, they were bilaterally microinjected into the dorsal CA1 area by amyloid-beta (Aß)25-35 for four consecutive days. Seven days after the last injection, they were trained to acquire Morris Water Maze (MWM) task for three successive days when they were treated with SP after each training session. In the second experiment, SP was administered 30 min after the first Aß microinjection and behavioral tests were performed one week after the last Aß administration. The activity of glutamate dehydrogenase (GDH), and glutamine synthetase (GS), as key enzymes involved in glutamate-glutamine homeostasis and histological assays were evaluated in the hippocampi. RESULTS: Our behavioral results indicated that post-training SP treatment enhanced task acquisition but did not change memory performance in Aß-treated rats. However, administration of SP at the time of Aß injection precludes the deteriorative effect of Aß and neuronal injury on both spatial learning and memory performances indicating its preventive action against Aß pathology at its early stages. Measurement of enzymes activity shows that α-KGDHC activity was reduced in the Aß treated group, and SP administration restored its activity; also, GDH and GS activities were increased and decreased respectively due to Aß, and SP reversed the action of Aß on these enzymes. CONCLUSIONS: This study proposes that SP possibly a promising therapeutic approach to improve memory impairment in AD, especially in the early phases of this disease.


Sujet(s)
Maladie d'Alzheimer , Phosphonates , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Animaux , Modèles animaux de maladie humaine , Glutamate dehydrogenase/métabolisme , Glutamate dehydrogenase/pharmacologie , Glutamate dehydrogenase/usage thérapeutique , Glutamate-ammonia ligase/métabolisme , Glutamate-ammonia ligase/pharmacologie , Glutamates/pharmacologie , Glutamine/métabolisme , Glutamine/pharmacologie , Hippocampe/métabolisme , Homéostasie , Ketoglutarate dehydrogenase complex/métabolisme , Ketoglutarate dehydrogenase complex/pharmacologie , Acides cétoglutariques/métabolisme , Acides cétoglutariques/pharmacologie , Acides cétoglutariques/usage thérapeutique , Mâle , Apprentissage du labyrinthe , Troubles de la mémoire/traitement médicamenteux , Troubles de la mémoire/métabolisme , Troubles de la mémoire/prévention et contrôle , Phosphonates/métabolisme , Phosphonates/pharmacologie , Phosphonates/usage thérapeutique , Fragments peptidiques/métabolisme , Fragments peptidiques/pharmacologie , Rats , Rat Wistar
11.
Trends Endocrinol Metab ; 33(2): 136-146, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34952764

RÉSUMÉ

Alpha-ketoglutarate (AKG) is an intermediate in the Krebs cycle involved in various metabolic and cellular pathways. As an antioxidant, AKG interferes in nitrogen and ammonia balance, and affects epigenetic and immune regulation. These pleiotropic functions of AKG suggest it may also extend human healthspan. Recent studies in worms and mice support this concept. A few studies published in the 1980s and 1990s in humans suggested the potential benefits of AKG in muscle growth, wound healing, and in promoting faster recovery after surgery. So far there are no recently published studies demonstrating the role of AKG in treating aging and age-related diseases; hence, further clinical studies are required to better understand the role of AKG in humans. This review will discuss the regulatory role of AKG in aging, as well as its potential therapeutic use in humans to treat age-related diseases.


Sujet(s)
Antioxydants , Acides cétoglutariques , Animaux , Antioxydants/métabolisme , Cycle citrique , Compléments alimentaires , Humains , Acides cétoglutariques/métabolisme , Acides cétoglutariques/usage thérapeutique , Souris
12.
EBioMedicine ; 73: 103672, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34740102

RÉSUMÉ

BACKGROUND: Phospho-Akt1 (pAkt1) undergoes prolyl hydroxylation at Pro125 and Pro313 by the prolyl hydroxylase-2 (PHD2) in a reaction decarboxylating α-ketoglutarate (αKG). We investigated whether the αKG supplementation could inhibit Akt-mediated activation of platelets and monocytes, in vitro as well as in vivo, by augmenting PHD2 activity. METHODS: We treated platelets or monocytes isolated from healthy individuals with αKG in presence of agonists in vitro and assessed the signalling molecules including pAkt1. We supplemented mice with dietary αKG and estimated the functional responses of platelets and monocytes ex vivo. Further, we investigated the impact of dietary αKG on inflammation and thrombosis in lungs of mice either treated with thrombosis-inducing agent carrageenan or infected with SARS-CoV-2. FINDINGS: Octyl αKG supplementation to platelets promoted PHD2 activity through elevated intracellular αKG to succinate ratio, and reduced aggregation in vitro by suppressing pAkt1(Thr308). Augmented PHD2 activity was confirmed by increased hydroxylated-proline and enhanced binding of PHD2 to pAkt in αKG-treated platelets. Contrastingly, inhibitors of PHD2 significantly increased pAkt1 in platelets. Octyl-αKG followed similar mechanism in monocytes to inhibit cytokine secretion in vitro. Our data also describe a suppressed pAkt1 and reduced activation of platelets and leukocytes ex vivo from mice supplemented with dietary αKG, unaccompanied by alteration in their number. Dietary αKG significantly reduced clot formation and leukocyte accumulation in various organs including lungs of mice treated with thrombosis-inducing agent carrageenan. Importantly, in SARS-CoV-2 infected hamsters, we observed a significant rescue effect of dietary αKG on inflamed lungs with significantly reduced leukocyte accumulation, clot formation and viral load alongside down-modulation of pAkt in the lung of the infected animals. INTERPRETATION: Our study suggests that dietary αKG supplementation prevents Akt-driven maladies such as thrombosis and inflammation and rescues pathology of COVID19-infected lungs. FUNDING: Study was funded by the Department of Biotechnology (DBT), Govt. of India (grants: BT/PR22881 and BT/PR22985); and the Science and Engineering Research Board, Govt. of India (CRG/000092).


Sujet(s)
Acides cétoglutariques/usage thérapeutique , Prolyl hydroxylases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Thrombose/prévention et contrôle , Animaux , Plaquettes/cytologie , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , COVID-19/anatomopathologie , COVID-19/prévention et contrôle , COVID-19/médecine vétérinaire , COVID-19/virologie , Cricetinae , Compléments alimentaires , Régulation négative/effets des médicaments et des substances chimiques , Humains , Acides cétoglutariques/pharmacologie , Poumon/métabolisme , Poumon/anatomopathologie , Mesocricetus , Souris , Souris de lignée BALB C , Monocytes/cytologie , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Phosphorylation , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Protéines proto-oncogènes c-akt/génétique , SARS-CoV-2/isolement et purification , SARS-CoV-2/physiologie , Thrombose/induit chimiquement , Thrombose/anatomopathologie , Thrombose/médecine vétérinaire
13.
Food Funct ; 12(14): 6214-6225, 2021 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-34105579

RÉSUMÉ

Obesity is an issue of great concern to people all over the world. It is accompanied by serious complications, leading to reduced quality of life and higher morbidity and mortality. Over the past few years, there has been an explosion in knowledge about the roles of potential therapeutic agents in obesity management. Among them, amino acid (AA) derivatives, such as taurine, glutathione (GSH), betaine, α-ketoglutarate (AKG), ß-aminoisobutyric acid (BAIBA), and ß-hydroxy-ß-methylbutyrate (HMB), have recently gained popularity due to their beneficial effects on the promotion of weight loss and improvement in the lipid profile. The mechanisms of action of these derivatives mainly include inhibiting adipogenesis, increasing lipolysis, promoting brown/beige adipose tissue (BAT) development, and improving glucose metabolism. Therefore, this review summarizes these AA derivatives and the possible mechanisms responsible for their anti-obesity effects. Based on the current findings, these AA derivatives could be potential therapeutic agents for obesity and its related metabolic diseases.


Sujet(s)
Acides aminés/métabolisme , Acides aminés/pharmacologie , Obésité/traitement médicamenteux , Adipogenèse/effets des médicaments et des substances chimiques , Tissu adipeux beige/métabolisme , Tissu adipeux brun/métabolisme , Acides aminés/usage thérapeutique , Acides amino-isobutyriques/usage thérapeutique , Animaux , Bétaïne/usage thérapeutique , Glucose/métabolisme , Glutathion/usage thérapeutique , Humains , Acides cétoglutariques/usage thérapeutique , Lipolyse/effets des médicaments et des substances chimiques , Obésité/métabolisme , Taurine/usage thérapeutique , Valérates/usage thérapeutique , Perte de poids
14.
Nat Commun ; 11(1): 5596, 2020 11 05.
Article de Anglais | MEDLINE | ID: mdl-33154378

RÉSUMÉ

Age-related osteoporosis is characterized by the deterioration in bone volume and strength, partly due to the dysfunction of bone marrow mesenchymal stromal/stem cells (MSCs) during aging. Alpha-ketoglutarate (αKG) is an essential intermediate in the tricarboxylic acid (TCA) cycle. Studies have revealed that αKG extends the lifespan of worms and maintains the pluripotency of embryonic stem cells (ESCs). Here, we show that the administration of αKG increases the bone mass of aged mice, attenuates age-related bone loss, and accelerates bone regeneration of aged rodents. αKG ameliorates the senescence-associated (SA) phenotypes of bone marrow MSCs derived from aged mice, as well as promoting their proliferation, colony formation, migration, and osteogenic potential. Mechanistically, αKG decreases the accumulations of H3K9me3 and H3K27me3, and subsequently upregulates BMP signaling and Nanog expression. Collectively, our findings illuminate the role of αKG in rejuvenating MSCs and ameliorating age-related osteoporosis, with a promising therapeutic potential in age-related diseases.


Sujet(s)
Vieillissement , Histone/métabolisme , Acides cétoglutariques/usage thérapeutique , Ostéoporose/traitement médicamenteux , Vieillissement/métabolisme , Vieillissement/anatomopathologie , Animaux , Marqueurs biologiques/métabolisme , Protéines morphogénétiques osseuses/métabolisme , Régénération osseuse/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Femelle , Acides cétoglutariques/sang , Mâle , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Méthylation/effets des médicaments et des substances chimiques , Souris , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéoporose/métabolisme , Ostéoporose/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques
15.
J Cardiothorac Vasc Anesth ; 34(4): 959-965, 2020 Apr.
Article de Anglais | MEDLINE | ID: mdl-31543295

RÉSUMÉ

OBJECTIVE: More than 30% of European pediatric cardiac surgery centers use single-dose cold histidine-tryptophan-ketoglutarate cardioplegia (Custodiol; Dr Franz Köhler Chemie GmbH, Bensheim, Germany). In neonates with transposition of the great arteries, arterial switch surgery (ASO) implies aortic division, and it is unknown whether repeated ostial cannulation causes intimal insult and affects long-term results, and therefore, single-dose Custodiol is appealing. The present study investigated the association among myocardial no-flow duration, postoperative troponins, and postoperative outcomes in neonates undergoing ASO with Custodiol cardioplegia. DESIGN: Retrospective analysis of the association among myocardial no-flow duration, postoperative troponin release (concentration magnitude × measurement duration within 48 h), and outcomes using stratification according to coronary anatomy and attending surgeon. SETTING: Single-institutional, tertiary pediatric cardiac surgery unit of a university hospital. PARTICIPANTS: The study comprised 101 neonates undergoing ASO. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: The mean age of patients was 6.1 ± 5.4 days, the cardiopulmonary bypass duration was 108.7 ± 54.1 minutes, the temperature during cross-clamping was 31.1°C ± 1.7°C, the duration of mechanical ventilation was 4 (3-6) days, the length of intensive care unit stay was 7 (5-8) days, delayed sternal closure occurred in 32 (31.7%) patients, and no patients died. The myocardial no-flow duration averaged 62.3 ± 14.6 minutes and was linked with both troponin release (p = 0.04) and low cardiac output syndrome, as assessed by the requirement for delayed sternal closure (p = 0.03), regardless of cardiopulmonary bypass duration and temperature. Eighty-two percent of the patients with myocardial no-flow duration >74 minutes necessitated delayed sternal closure. CONCLUSIONS: Single-dose Custodiol may be inadequate for prolonged cross-clamping durations without myocardial perfusion in neonates.


Sujet(s)
Solutions cardioplégiques/usage thérapeutique , Histidine/usage thérapeutique , Acides cétoglutariques/usage thérapeutique , Transposition des gros vaisseaux/chirurgie , Tryptophane/usage thérapeutique , Constriction , Allemagne , Arrêt cardiaque provoqué , Humains , Nouveau-né , Études rétrospectives
16.
Drug Chem Toxicol ; 43(5): 546-551, 2020 Sep.
Article de Anglais | MEDLINE | ID: mdl-31094237

RÉSUMÉ

Alcohol is the most abused psychoactive substance and known hepatotoxicant. Present study elucidates possible therapeutic effect of oral alpha-ketoglutarate (AKG) supplementation against alcohol induced hepatic dysfunction, using biochemical, histopathological and most importantly, in vivo functional imaging approaches. Animals were divided into three groups of 6 animals each. Group-I (control): Normal saline; Group-II: 20% (v/v) solution of ethanol (5 ml/day) intragastrically using oral gavage for 2 months. Group-III: ethanol treatment as in group-II along with AKG supplementation (2g/kg/bw; intragastrically using oral gavage for 2 months). In vivo hepatobiliary scintigraphy was performed in all animals using 99mTc-mebrofenin (99mTc-MEB) as radiotracer to determine changes in (a) Hepatic extraction fraction (HEF), for quantification of radiotracer uptake, (b) Time to reach maximum hepatic uptake (Tpeak), and (c) Time for hepatic uptake to reduce by 50% (T1/2peak). Biochemical (alanine aminotransferase, aspartate aminotransferase, reduced glutathione, superoxide dismutase, catalase, and lipid peroxidation) and histological parameters were also studied. Hepatic uptake and excretion kinetics using 99mTc-MEB scintigraphy showed prompt 99mTc-MEB clearance from liver in control group (HEF: 91.26 ± 2.32; Tpeak: 143 ± 23 sec; T1/2peak: 434 ± 41 sec), while it was significantly abnormal in ethanol group and showed less efficient radiotracer accumulation (HEF: 62.72 ± 5.6; Tpeak: 201 ± 33 sec; T1/2peak: 542 ± 52 sec). Supplementation of AKG along with ethanol significantly improved liver function (HEF: 76.42 ± 5.3; Tpeak: 155 ± 34 sec; T1/2peak: 455 ± 22 sec). Biochemical and histopathology parameters were correlative to findings of functional imaging study. Results strongly indicate hepatoprotective potential of AKG against alcohol-induced hepatic injury. Study further proposes the use of in vivo hepatobiliary scintigraphy for high throughput screening of other hepatoprotectants.


Sujet(s)
Éthanol/toxicité , Acides cétoglutariques/usage thérapeutique , Maladies alcooliques du foie/traitement médicamenteux , Maladies du foie/prévention et contrôle , Foie/effets des médicaments et des substances chimiques , Alanine transaminase/analyse , Animaux , Aspartate aminotransferases/analyse , Modèles animaux de maladie humaine , Acides cétoglutariques/pharmacologie , Foie/enzymologie , Maladies alcooliques du foie/enzymologie , Mâle , Scintigraphie , Rats , Rat Sprague-Dawley
17.
Aging Cell ; 19(1): e13059, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31691468

RÉSUMÉ

Aging usually involves the progressive development of certain illnesses, including diabetes and obesity. Due to incapacity to form new white adipocytes, adipose expansion in aged mice primarily depends on adipocyte hypertrophy, which induces metabolic dysfunction. On the other hand, brown adipose tissue burns fatty acids, preventing ectopic lipid accumulation and metabolic diseases. However, the capacity of brown/beige adipogenesis declines inevitably during the aging process. Previously, we reported that DNA demethylation in the Prdm16 promoter is required for beige adipogenesis. DNA methylation is mediated by ten-eleven family proteins (TET) using alpha-ketoglutarate (AKG) as a cofactor. Here, we demonstrated that the circulatory AKG concentration was reduced in middle-aged mice (10-month-old) compared with young mice (2-month-old). Through AKG administration replenishing the AKG pool, aged mice were associated with the lower body weight gain and fat mass, and improved glucose tolerance after challenged with high-fat diet (HFD). These metabolic changes are accompanied by increased expression of brown adipose genes and proteins in inguinal adipose tissue. Cold-induced brown/beige adipogenesis was impeded in HFD mice, whereas AKG rescued the impairment of beige adipocyte functionality in middle-aged mice. Besides, AKG administration up-regulated Prdm16 expression, which was correlated with an increase of DNA demethylation in the Prdm16 promoter. In summary, AKG supplementation promotes beige adipogenesis and alleviates HFD-induced obesity in middle-aged mice, which is associated with enhanced DNA demethylation of the Prdm16 gene.


Sujet(s)
Adipogenèse/effets des médicaments et des substances chimiques , Acides cétoglutariques/usage thérapeutique , Obésité/traitement médicamenteux , Animaux , Femelle , Acides cétoglutariques/pharmacologie , Souris , Obésité/prévention et contrôle
18.
Cell Metab ; 27(5): 1007-1025.e5, 2018 May 01.
Article de Anglais | MEDLINE | ID: mdl-29657030

RÉSUMÉ

Using molecular, biochemical, and untargeted stable isotope tracing approaches, we identify a previously unappreciated glutamine-derived α-ketoglutarate (αKG) energy-generating anaplerotic flux to be critical in mitochondrial DNA (mtDNA) mutant cells that harbor human disease-associated oxidative phosphorylation defects. Stimulating this flux with αKG supplementation enables the survival of diverse mtDNA mutant cells under otherwise lethal obligatory oxidative conditions. Strikingly, we demonstrate that when residual mitochondrial respiration in mtDNA mutant cells exceeds 45% of control levels, αKG oxidative flux prevails over reductive carboxylation. Furthermore, in a mouse model of mitochondrial myopathy, we show that increased oxidative αKG flux in muscle arises from enhanced alanine synthesis and release into blood, concomitant with accelerated amino acid catabolism from protein breakdown. Importantly, in this mouse model of mitochondriopathy, muscle amino acid imbalance is normalized by αKG supplementation. Taken together, our findings provide a rationale for αKG supplementation as a therapeutic strategy for mitochondrial myopathies.


Sujet(s)
ADN mitochondrial/génétique , Glutamine/métabolisme , Acides cétoglutariques , Mitochondries , Myopathies mitochondriales , Adaptation physiologique , Alanine/métabolisme , Animaux , Modèles animaux de maladie humaine , Métabolisme énergétique , Cellules HeLa , Humains , Acides cétoglutariques/métabolisme , Acides cétoglutariques/usage thérapeutique , Mâle , Souris , Mitochondries/génétique , Mitochondries/métabolisme , Myopathies mitochondriales/génétique , Myopathies mitochondriales/métabolisme , Mutation , Phosphorylation oxydative
19.
FASEB J ; 32(1): 488-499, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-28939592

RÉSUMÉ

Skeletal muscle atrophy due to excessive protein degradation is the main cause for muscle dysfunction, fatigue, and weakening of athletic ability. Endurance exercise is effective to attenuate muscle atrophy, but the underlying mechanism has not been fully investigated. α-Ketoglutarate (AKG) is a key intermediate of tricarboxylic acid cycle, which is generated during endurance exercise. Here, we demonstrated that AKG effectively attenuated corticosterone-induced protein degradation and rescued the muscle atrophy and dysfunction in a Duchenne muscular dystrophy mouse model. Interestingly, AKG also inhibited the expression of proline hydroxylase 3 (PHD3), one of the important oxidoreductases expressed under hypoxic conditions. Subsequently, we identified the ß2 adrenergic receptor (ADRB2) as a downstream target for PHD3. We found AKG inhibited PHD3/ADRB2 interaction and therefore increased the stability of ADRB2. In addition, combining pharmacologic and genetic approaches, we showed that AKG rescues skeletal muscle atrophy and protein degradation through a PHD3/ADRB2 mediated mechanism. Taken together, these data reveal a mechanism for inhibitory effects of AKG on muscle atrophy and protein degradation. These findings not only provide a molecular basis for the potential use of exercise-generated metabolite AKG in muscle atrophy treatment, but also identify PHD3 as a potential target for the development of therapies for muscle wasting.-Cai, X., Yuan, Y., Liao, Z., Xing, K., Zhu, C., Xu, Y., Yu, L., Wang, L., Wang, S., Zhu, X., Gao, P., Zhang, Y., Jiang, Q., Xu, P., Shu, G. α-Ketoglutarate prevents skeletal muscle protein degradation and muscle atrophy through PHD3/ADRB2 pathway.


Sujet(s)
Acides cétoglutariques/usage thérapeutique , Protéines du muscle/métabolisme , Myopathie de Duchenne/traitement médicamenteux , Procollagen-Proline Dioxygenase/métabolisme , Récepteurs bêta-2 adrénergiques/métabolisme , Animaux , Corticostérone/pharmacologie , Modèles animaux de maladie humaine , Mâle , Voies et réseaux métaboliques/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Souris de lignée mdx , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/anatomopathologie , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/prévention et contrôle , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Stabilité protéique/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques
20.
Arch Immunol Ther Exp (Warsz) ; 65(1): 21-36, 2017 Feb.
Article de Anglais | MEDLINE | ID: mdl-27326424

RÉSUMÉ

Alpha-ketoglutarate (AKG), an endogenous intermediary metabolite in the Krebs cycle, is a molecule involved in multiple metabolic and cellular pathways. It functions as an energy donor, a precursor in the amino acid biosynthesis, a signalling molecule, as well as a regulator of epigenetic processes and cellular signalling via protein binding. AKG is an obligatory co-substrate for 2-oxoglutarate-dependent dioxygenases, which catalyse hydroxylation reactions on various types of substrates. It regulates the activity of prolyl-4 hydroxylase, which controls the biosynthesis of collagen, a component of bone tissue. AKG also affects the functioning of prolyl hydroxylases, which, in turn, influences the function of the hypoxia-inducible factor, an important transcription factor in cancer development and progression. Additionally, it affects the functioning of enzymes that influence epigenetic modifications of chromatin: ten-eleven translocation hydroxylases involved in DNA demethylation and the Jumonji C domain containing lysine demethylases, which are the major histone demethylases. Thus, it regulates gene expression. The metabolic and extrametabolic function of AKG in cells and the organism open many different fields for therapeutic interventions for treatment of diseases. This review presents the results of studies conducted with the use of AKG in states of protein deficiency and oxidative stress conditions. It also discusses current knowledge about AKG as an immunomodulatory agent and a bone anabolic factor. Additionally, the regulatory role of AKG and its structural analogues in carcinogenesis as well as the results of studies of AKG as an anticancer agent are discussed.


Sujet(s)
Acides cétoglutariques/usage thérapeutique , Anabolisants/composition chimique , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/immunologie , Antioxydants/composition chimique , Cycle citrique , Méthylation de l'ADN , Compléments alimentaires , Épigenèse génétique , Humains , Hypoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie/composition chimique , Mutation , Stress oxydatif , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...