Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Publication year range
1.
Adv Nutr ; 15(4): 100200, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38438107

ABSTRACT

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.


Subject(s)
Inflammatory Bowel Diseases , Ketoglutaric Acids , Humans , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/prevention & control , Intestinal Mucosa
2.
Mol Med ; 30(1): 15, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38254035

ABSTRACT

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.


Subject(s)
Aortic Valve Stenosis , Ferroptosis , Heart Failure , Ketoglutaric Acids , Mitophagy , Angiotensin II , Chromatography, Liquid , Ferroptosis/drug effects , Fibrosis , Heart Failure/drug therapy , Heart Failure/metabolism , Hypertrophy , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/therapeutic use , Mitophagy/drug effects , Myocytes, Cardiac , NAD , Protein Kinases , RNA, Small Interfering , Sirtuin 1 , Tandem Mass Spectrometry , Animals , Mice
3.
Acta Biomater ; 173: 442-456, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37984632

ABSTRACT

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.


Subject(s)
Ketoglutaric Acids , Osteoporosis , Humans , Mice , Animals , Aged , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/therapeutic use , Osteogenesis , Luminescence , Osteoporosis/drug therapy , Osteoblasts/metabolism
4.
Redox Biol ; 62: 102663, 2023 06.
Article in English | MEDLINE | ID: mdl-36924682

ABSTRACT

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.


Subject(s)
Ketoglutaric Acids , Osteoarthritis , Humans , Rats , Animals , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/therapeutic use , Mitophagy , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Chondrocytes/metabolism , Oxidative Stress , Interleukin-1beta/metabolism , Cells, Cultured
5.
Cell Death Dis ; 14(2): 170, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36854755

ABSTRACT

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.


Subject(s)
B7-H1 Antigen , Ketoglutaric Acids , Melanoma , Animals , Mice , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Dietary Supplements , Epigenesis, Genetic , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/therapeutic use , Melanoma/drug therapy , Melanoma/genetics
6.
Biomater Sci ; 10(23): 6688-6697, 2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36190458

ABSTRACT

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.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Nanoparticles , Mice , Animals , Methotrexate/pharmacology , Methotrexate/therapeutic use , Ketoglutaric Acids/therapeutic use , Mice, Inbred DBA , Arthritis, Rheumatoid/drug therapy , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , T-Lymphocytes, Helper-Inducer/metabolism , Polymers/therapeutic use
7.
Trends Endocrinol Metab ; 33(2): 136-146, 2022 02.
Article in English | MEDLINE | ID: mdl-34952764

ABSTRACT

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.


Subject(s)
Antioxidants , Ketoglutaric Acids , Animals , Antioxidants/metabolism , Citric Acid Cycle , Dietary Supplements , Humans , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/therapeutic use , Mice
8.
EBioMedicine ; 73: 103672, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34740102

ABSTRACT

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).


Subject(s)
Ketoglutaric Acids/therapeutic use , Prolyl Hydroxylases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thrombosis/prevention & control , Animals , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , COVID-19/pathology , COVID-19/prevention & control , COVID-19/veterinary , COVID-19/virology , Cricetinae , Dietary Supplements , Down-Regulation/drug effects , Humans , Ketoglutaric Acids/pharmacology , Lung/metabolism , Lung/pathology , Mesocricetus , Mice , Mice, Inbred BALB C , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation , Platelet Aggregation/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/genetics , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Thrombosis/chemically induced , Thrombosis/pathology , Thrombosis/veterinary
9.
Drug Chem Toxicol ; 43(5): 546-551, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31094237

ABSTRACT

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.


Subject(s)
Ethanol/toxicity , Ketoglutaric Acids/therapeutic use , Liver Diseases, Alcoholic/drug therapy , Liver Diseases/prevention & control , Liver/drug effects , Alanine Transaminase/analysis , Animals , Aspartate Aminotransferases/analysis , Disease Models, Animal , Ketoglutaric Acids/pharmacology , Liver/enzymology , Liver Diseases, Alcoholic/enzymology , Male , Radionuclide Imaging , Rats , Rats, Sprague-Dawley
10.
Aging Cell ; 19(1): e13059, 2020 01.
Article in English | MEDLINE | ID: mdl-31691468

ABSTRACT

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.


Subject(s)
Adipogenesis/drug effects , Ketoglutaric Acids/therapeutic use , Obesity/drug therapy , Animals , Female , Ketoglutaric Acids/pharmacology , Mice , Obesity/prevention & control
11.
Cell Metab ; 27(5): 1007-1025.e5, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29657030

ABSTRACT

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.


Subject(s)
DNA, Mitochondrial/genetics , Glutamine/metabolism , Ketoglutaric Acids , Mitochondria , Mitochondrial Myopathies , Adaptation, Physiological , Alanine/metabolism , Animals , Disease Models, Animal , Energy Metabolism , HeLa Cells , Humans , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/therapeutic use , Male , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/metabolism , Mutation , Oxidative Phosphorylation
12.
Arch Immunol Ther Exp (Warsz) ; 65(1): 21-36, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27326424

ABSTRACT

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.


Subject(s)
Ketoglutaric Acids/therapeutic use , Anabolic Agents/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Antioxidants/chemistry , Citric Acid Cycle , DNA Methylation , Dietary Supplements , Epigenesis, Genetic , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Mutation , Oxidative Stress , Signal Transduction
13.
J Vet Med Sci ; 78(4): 563-71, 2016 May 03.
Article in English | MEDLINE | ID: mdl-26725871

ABSTRACT

The aim of the study was to investigate the effect of 2-oxoglutaric acid (2-Ox) supplementation (a precursor of glutamine and hydroxyproline, the most abundant amino acid of collagen) on cartilage and bone in pigs after fundectomy. Pigs at the age of forty days were subjected to fundectomy and divided into two groups depending on 2-Ox supplementation (at the daily dosage of 0.4 g/kg of body weight). Other pigs were sham operated. Pigs were euthanized at the age of eight months. An analysis of the morphometry of trabeculae, growth plate and articular cartilage in fundectomy-induced osteopenic bone was performed. Moreover, the levels of expression of osteocalcin, osteopontin and osteoprotegerin in trabecular bone and osteocalcin in articular cartilage were evaluated. Articular cartilage was thinnest in fundectomized pigs and thickest in 2-Ox-supplemented animals after fundectomy. Moreover, 2-Ox supplementation after fundectomy enhanced the total thickness of the growth plate and trabeculae in fundectomized pigs. The most evident signal for osteocalcin and osteoprotegerin in trabecular bone was in sham-operated and 2-Ox-supplemented pigs; a low reaction was observed in the fundectomized group. Additionally, as a long-term postoperative consequence, a change was observed in the expression of osteocalcin in articular cartilage. It seems that 2-Ox is suitable for use in preventing the negative effects of fundectomy on cancellous bone and cartilage.


Subject(s)
Bone Diseases, Metabolic/diet therapy , Cancellous Bone , Cartilage, Articular , Growth Plate , Ketoglutaric Acids/therapeutic use , Animals , Body Weight , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/pathology , Cancellous Bone/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Collagen/metabolism , Gastric Fundus/surgery , Growth Plate/pathology , Male , Osteocalcin/biosynthesis , Proteoglycans/metabolism , Swine
14.
PLoS One ; 9(7): e103526, 2014.
Article in English | MEDLINE | ID: mdl-25061944

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS), also known as Lou Gehrig's disease, is a neurodegenerative disorder of motor neurons causing progressive muscle weakness, paralysis, and eventual death from respiratory failure. There is currently no cure or effective treatment for ALS. Besides motor neuron degeneration, ALS is associated with impaired energy metabolism, which is pathophysiologically linked to mitochondrial dysfunction and glutamate excitotoxicity. The Deanna Protocol (DP) is a metabolic therapy that has been reported to alleviate symptoms in patients with ALS. In this study we hypothesized that alternative fuels in the form of TCA cycle intermediates, specifically arginine-alpha-ketoglutarate (AAKG), the main ingredient of the DP, and the ketogenic diet (KD), would increase motor function and survival in a mouse model of ALS (SOD1-G93A). ALS mice were fed standard rodent diet (SD), KD, or either diets containing a metabolic therapy of the primary ingredients of the DP consisting of AAKG, gamma-aminobutyric acid, Coenzyme Q10, and medium chain triglyceride high in caprylic triglyceride. Assessment of ALS-like pathology was performed using a pre-defined criteria for neurological score, accelerated rotarod test, paw grip endurance test, and grip strength test. Blood glucose, blood beta-hydroxybutyrate, and body weight were also monitored. SD+DP-fed mice exhibited improved neurological score from age 116 to 136 days compared to control mice. KD-fed mice exhibited better motor performance on all motor function tests at 15 and 16 weeks of age compared to controls. SD+DP and KD+DP therapies significantly extended survival time of SOD1-G93A mice by 7.5% (p = 0.001) and 4.2% (p = 0.006), respectively. Sixty-three percent of mice in the KD+DP and 72.7% of the SD+DP group lived past 125 days, while only 9% of the control animals survived past that point. Targeting energy metabolism with metabolic therapy produces a therapeutic effect in ALS mice which may prolong survival and quality of life in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Arginine/analogs & derivatives , Caprylates/therapeutic use , Ketoglutaric Acids/therapeutic use , Ubiquinone/analogs & derivatives , gamma-Aminobutyric Acid/therapeutic use , Amyotrophic Lateral Sclerosis/genetics , Animals , Arginine/administration & dosage , Arginine/therapeutic use , Caprylates/administration & dosage , Dietary Supplements , Ketoglutaric Acids/administration & dosage , Male , Mice , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Ubiquinone/administration & dosage , Ubiquinone/therapeutic use , gamma-Aminobutyric Acid/administration & dosage
15.
Curr Med Res Opin ; 28(11): 1767-74, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23043451

ABSTRACT

OBJECTIVE: Arthroscopic rotator cuff repair generally provides satisfactory result, in terms of decreasing shoulder pain, resulting in improvement in range of motion. Unfortunately, imaging studies have shown that after surgical repair re-rupture rate is potentially high. Literature data indicate that each of the components present in a commercial supplement sold in Italy as Tenosan * (arginine L-alpha-ketoglutarate, methylsulfonylmethane, hydrolyzed type I collagen and bromelain) have a potential role in tendon healing and mitigating the pain due to tendonitis. We evaluated the clinical and MRI results of rotator cuff repair with and without the employment of this oral supplement in patients with a large, postero-superior rotator cuff tear (RCT). RESEARCH DESIGN AND METHODS: We enrolled 90 consecutive patients who had a large, postero-superior RCT. All the lesions were managed with an arthroscopic repair. Patients were randomized and treated either with (Group I) or without (Group II) the supplement. The primary outcomes were the difference between the pre- and post-operative Constant score and repair integrity assessed by MRI according to Sugaya's classification. The secondary outcome was the pre- and post-operative Simple Shoulder Test. RESULTS: No statistically significant differences were identified between the two groups for each considered variable, except for shoulder pain (follow-up: 6 months) and repair integrity (final follow-up). Intensity of shoulder pain was lower in the Group I patients (p < 0.001). Analogously, in Group I, the percentage of patients with a better repair integrity result was significantly higher than Group II. CONCLUSION: The use of the supplement for 3 months after cuff repair decreases shoulder post-operative pain and leads to a slight improvement in repair integrity. This improvement does not seem to correlate with an better objective functional outcome. However, these effects could facilitate and abbreviate the post-operative rehabilitation program and reduce re-rupture rate. The main limitations of this study are the relative short follow-up period and small number of patients studied.


Subject(s)
Arginine/analogs & derivatives , Bromelains/pharmacology , Collagen Type I/pharmacology , Dimethyl Sulfoxide/pharmacology , Ketoglutaric Acids/pharmacology , Rotator Cuff Injuries , Shoulder Impingement Syndrome/drug therapy , Sulfones/pharmacology , Wound Healing/drug effects , Aged , Arginine/pharmacology , Arginine/therapeutic use , Bromelains/therapeutic use , Collagen Type I/therapeutic use , Dimethyl Sulfoxide/therapeutic use , Female , Humans , Hydrolysis , Ketoglutaric Acids/therapeutic use , Male , Middle Aged , Prospective Studies , Sulfones/therapeutic use
16.
Am J Physiol Renal Physiol ; 303(1): F56-63, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22513847

ABSTRACT

Acute kidney injury (AKI) is the most common kidney disease in hospitalized patients with high mortality. Ischemia and reperfusion (I/R) is one of the major causes of AKI. The combination of α-ketoglutarate+malate (αKG/MAL) showed the ability to reduce hypoxia-induced damage to isolated proximal tubules. The present study utilizes a rat model of I/R-induced AKI accompanied by intensive biomonitoring to examine whether αKG/MAL provides protection in vivo. AKI was induced in male Sprague-Dawley rats by bilateral renal clamping (40 min) followed by reperfusion (240 min). αKG/MAL was infused continuously for 60 min before and 45 min after ischemia. Normoxic and I/R control groups received 0.9% NaCl solution. The effect of αKG/MAL was evaluated by biomonitoring, blood and plasma parameters, histopathology, and immunohistochemical staining for kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL), as well as by determination of tissue ATP and nonesterified fatty acid concentrations. Intravenous infusion of αKG/MAL at a cumulative dose of 1 mmol/kg each (146 mg/kg αKG and 134 mg/kg MAL) did not prevent I/R-induced increases in plasma creatinine, histopathological alterations, or cortical ATP depletion. On the contrary, the most notable adverse affect in animals receiving αKG/MAL was the decrease in mean arterial blood pressure, which was also accompanied by a reduction in heart rate. Supplementation with αKG/MAL, which is very protective against hypoxia-induced injury in isolated proximal tubules, does not protect against I/R-induced renal injury in vivo, possibly due to cardiovascular depressive effects.


Subject(s)
Acute Kidney Injury/pathology , Blood Pressure/drug effects , Heart Rate/drug effects , Ketoglutaric Acids/toxicity , Kidney/drug effects , Malates/toxicity , Reperfusion Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Animals , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia/physiopathology , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/therapeutic use , Kidney/blood supply , Kidney/metabolism , Kidney/pathology , Malates/pharmacology , Malates/therapeutic use , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control
17.
Nutrition ; 28(2): 190-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22018909

ABSTRACT

OBJECTIVE: The potential effects of the prenatal administration of dexamethasone and the postnatal treatment with 2-oxoglutaric acid on postnatal development of the small intestine of farm animals have not been examined experimentally. The aim of this study was to establish the changes in morphologic parameters of the small intestine damaged by the prenatal action of dexamethasone in piglets supplemented with 2-oxoglutaric acid. METHODS: Three milligrams dexamethasone was administered intramuscularly every second day from day 70 of pregnancy to parturition and then piglets were supplemented with 2-oxoglutaric acid for 35 d of postnatal life (0.4 g/kg of body weight). The histomorphometry of the pig duodenum and jejunum was determined. Immunohistochemical staining with anti-Ki-67, CD3, null T cells, cadherin, claudin, and neurofilament antibodies was performed. RESULTS: Maternal treatment with dexamethasone decreased and limited the expression of claudin and cadherin in the epithelium. Dexamethasone led to thinning of the myenteron of the duodenum and the middle part of the jejunum in weaned piglets and influenced duodenal glands that became more elongated compared with control glands. Moreover, 2-oxoglutaric acid increased cell proliferation and the amount and maturity of peripheral blood lymphocytes in the duodenum and jejunum. It supported epithelial integrity and changed the circularity of the nerve plexuses. CONCLUSION: The 2-oxoglutaric acid administered to piglets while suckling induced a complete recovery from intestinal damage caused by the prenatal action of dexamethasone.


Subject(s)
Dexamethasone/adverse effects , Gastrointestinal Agents/therapeutic use , Glucocorticoids/adverse effects , Intestinal Diseases/prevention & control , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Ketoglutaric Acids/therapeutic use , Animals , Cadherins/metabolism , Cell Proliferation/drug effects , Claudins/metabolism , Disease Models, Animal , Enteric Nervous System/drug effects , Enteric Nervous System/pathology , Female , Gastrointestinal Agents/pharmacology , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Ketoglutaric Acids/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Pregnancy , Swine
19.
Br J Nutr ; 106(3): 357-63, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21342606

ABSTRACT

The present study determined whether α-ketoglutarate (AKG) might affect the expression of AMP-activated protein kinase (AMPK) and energy status in the intestinal mucosa of piglets challenged with Escherichia coli lipopolysaccharide (LPS). A total of eighteen piglets (weaned at 21 d of age) were allocated to one of three treatments: (1) non-challenged (control); (2) LPS-challenged (LPS); (3) LPS+1 % AKG (LPS+AKG). Piglets in the control and LPS groups were fed a maize- and soyabean meal-based diet, and the LPS+AKG group was fed the basal diet supplemented with 1 % AKG. On days 10, 12, 14 and 16 of the trial, piglets in the LPS and LPS+AKG groups were challenged with LPS (80 µg/kg body weight), whereas piglets in the control group received the same volume of sterile saline. Pigs were euthanised 24 h after the last administration of LPS or saline to obtain intestinal mucosae for biochemical analysis. Compared with the control group, LPS administration decreased (P < 0·05) the oxidation of AKG, oleic acid, glutamine and glucose in enterocytes, decreased concentrations of ATP in the duodenal and jejunal mucosae and decreased adenylate energy charge (AMP:ATP ratio) in the jejunal and ileal mucosae. Additionally, LPS treatment reduced (P < 0·05) mucosal concentrations of phosphorylated AMPK in the jejunum and ileum as well as acetyl-CoA carboxylase in all segments of the small intestine. The adverse effects of LPS were reversed by AKG. Collectively, these results indicate that dietary supplementation with 1 % AKG beneficially modulates the AMPK signalling pathway to improve energy status in the small intestine of LPS-challenged piglets.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Enterocytes/metabolism , Intestinal Diseases/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Ketoglutaric Acids/pharmacology , Acetyl-CoA Carboxylase/metabolism , Adenosine Monophosphate/metabolism , Animals , Dietary Supplements , Escherichia coli , Female , Glucose/metabolism , Glutamine/metabolism , Intestinal Diseases/chemically induced , Intestinal Diseases/drug therapy , Ketoglutaric Acids/therapeutic use , Lipopolysaccharides , Oleic Acid/metabolism , Oxidation-Reduction , Phosphorylation , Signal Transduction/drug effects , Swine , Weaning
20.
Amino Acids ; 39(2): 555-64, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20127262

ABSTRACT

Neonates are at increased risk for inflammatory bowel disease, but effective prevention and treatments are currently limited. This study was conducted with the lipopolysaccharide (LPS)-challenged piglet model to determine the effects of dietary supplementation with alpha-ketoglutarate (AKG) on the intestinal morphology and function. Eighteen 24-day-old pigs (weaned at 21 days of age) were assigned randomly to control, LPS, and LPS + AKG groups. The piglets in the control and LPS groups were fed a corn- and soybean meal-based diet, whereas the LPS + AKG group was fed the basal diet supplemented with 1% AKG. On days 10, 12, 14, and 16, piglets in the LPS and LPS + AKG groups received intraperitoneal administration of LPS (80 microg/kg BW), whereas piglets in the control group received the same volume of saline. On day 16, D-xylose was orally administrated to all pigs at the dose of 0.1 g/kg BW, 2 h after LPS or saline injection, and blood samples were collected 3 h thereafter. Twenty-four hours post-administration of LPS or saline, pigs were killed to obtain intestinal mucosae for analysis. Compared with the control group, LPS challenge reduced (P < 0.05) protein levels, the ratio of villus height to crypt depth, and the ratio of phosphorylated mTOR to total mTOR in duodenal, jejunal, and ileal mucosa. These adverse effects of LPS were attenuated (P < 0.05) by AKG supplementation. Moreover, AKG prevented the LPS-induced increase in intestinal HSP70 expression. Collectively, these novel results indicate that dietary supplementation with 1% AKG activates the mTOR signaling, alleviates the mucosal damage, and improves the absorptive function of the small intestine in LPS-challenged piglets. The findings not only help understand the mode of AKGs actions in the neonatal gut but also have important implications for infant nutrition under inflammatory conditions.


Subject(s)
Intestinal Mucosa/drug effects , Ketoglutaric Acids/pharmacology , Lipopolysaccharides/toxicity , Amino Acids/metabolism , Animals , Animals, Newborn , Dietary Supplements , Duodenum/chemistry , Eating/drug effects , HSP70 Heat-Shock Proteins/biosynthesis , Ileum/chemistry , Intestinal Diseases/chemically induced , Intestinal Diseases/prevention & control , Intestinal Mucosa/chemistry , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/physiology , Jejunum/chemistry , Ketoglutaric Acids/therapeutic use , Protein Serine-Threonine Kinases/physiology , Swine , TOR Serine-Threonine Kinases , Weaning , Weight Gain/drug effects , Xylose/blood
SELECTION OF CITATIONS
SEARCH DETAIL