Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cancer Chemother Pharmacol ; 93(1): 11-22, 2024 01.
Article in English | MEDLINE | ID: mdl-37728615

ABSTRACT

PURPOSE: Enhancing chemotherapy sensitivity in colorectal cancer (CRC) is critical for improving treatment outcomes. TMEM120A has been reported to interact with coenzyme A (CoA), but its biological significance in CRC is unknown. In this study, we aimed to investigate the functional implications of TMEM120A in CRC and its impact on chemotherapy sensitivity. METHODS: Stable knockout of TMEM120A in CRC cell lines was conducted using CRISPR/Cas9 technology. Overexpression of various derivatives of TMEM120A was achieved through lentiviral transduction. Cell fractionation was employed to isolate the nuclear and cytoplasmic fraction. Total histones were isolated by acid extraction and then subjected to determine histone acetylation levels using western blot analysis. Cell viability was evaluated using the MTS assay. RESULTS: We demonstrate that TMEM120A's nuclear localization is crucial for its role in regulating CRC chemosensitivity. Mechanistically, the nuclear subpopulation of TMEM120A plays a key role in sustaining the nuclear CoA levels, which in turn influences the levels of nuclear acetyl-CoA and histone acetylation in CRC cells. Notably, direct inhibition of histone acetylation recapitulated the phenotypic effects observed upon TMEM120A depletion, leading to increased chemosensitivity in CRC cells. CONCLUSION: Our study provides novel insights into the role of TMEM120A in modulating chemotherapy sensitivity in CRC. Nuclear TMEM120A regulates CoA levels, which in turn modulates nuclear acetyl-CoA levels and histone acetylation, thereby influencing the response of CRC cells to chemotherapy agents. Targeting TMEM120A-mediated pathways may represent a promising strategy for enhancing chemotherapy efficacy in CRC treatment.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Humans , Histones/metabolism , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/therapeutic use , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Acetylation
2.
J Pharmacol Exp Ther ; 388(1): 171-180, 2024 01 02.
Article in English | MEDLINE | ID: mdl-37875310

ABSTRACT

Pantothenate kinase-associated neurodegeneration (PKAN) is characterized by a motor disorder with combinations of dystonia, parkinsonism, and spasticity, leading to premature death. PKAN is caused by mutations in the PANK2 gene that result in loss or reduction of PANK2 protein function. PANK2 is one of three kinases that initiate and regulate coenzyme A biosynthesis from vitamin B5, and the ability of BBP-671, an allosteric activator of pantothenate kinases, to enter the brain and elevate coenzyme A was investigated. The metabolic stability, protein binding, and membrane permeability of BBP-671 all suggest that it has the physical properties required to cross the blood-brain barrier. BBP-671 was detected in plasma, liver, cerebrospinal fluid, and brain following oral administration in rodents, demonstrating the ability of BBP-671 to penetrate the brain. The pharmacokinetic and pharmacodynamic properties of orally administered BBP-671 evaluated in cannulated rats showed that coenzyme A (CoA) concentrations were elevated in blood, liver, and brain. BBP-671 elevation of whole-blood acetyl-CoA served as a peripheral pharmacodynamic marker and provided a suitable method to assess target engagement. BBP-671 treatment elevated brain coenzyme A concentrations and improved movement and body weight in a PKAN mouse model. Thus, BBP-671 crosses the blood-brain barrier to correct the brain CoA deficiency in a PKAN mouse model, resulting in improved locomotion and survival and providing a preclinical foundation for the development of BBP-671 as a potential treatment of PKAN. SIGNIFICANCE STATEMENT: The blood-brain barrier represents a major hurdle for drugs targeting brain metabolism. This work describes the pharmacokinetic/pharmacodynamic properties of BBP-671, a pantothenate kinase activator. BBP-671 crosses the blood-brain barrier to correct the neuron-specific coenzyme A (CoA) deficiency and improve motor function in a mouse model of pantothenate kinase-associated neurodegeneration. The central role of CoA and acetyl-CoA in intermediary metabolism suggests that pantothenate kinase activators may be useful in modifying neurological metabolic disorders.


Subject(s)
Pantothenate Kinase-Associated Neurodegeneration , Mice , Animals , Rats , Pantothenate Kinase-Associated Neurodegeneration/drug therapy , Pantothenate Kinase-Associated Neurodegeneration/genetics , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/therapeutic use , Coenzyme A/metabolism , Disease Models, Animal , Phosphotransferases (Alcohol Group Acceptor)/genetics , Brain/metabolism
3.
Acta Pharmacol Sin ; 45(2): 366-377, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37770579

ABSTRACT

Diabetic nephropathy (DN) is characterized by chronic low-grade renal inflammatory responses, which greatly contribute to disease progression. Abnormal glucose metabolism disrupts renal lipid metabolism, leading to lipid accumulation, nephrotoxicity, and subsequent aseptic renal interstitial inflammation. In this study, we investigated the mechanisms underlying the renal inflammation in diabetes, driven by glucose-lipid metabolic rearrangement with a focus on the role of acetyl-CoA synthetase 2 (ACSS2) in lipid accumulation and renal tubular injury. Diabetic models were established in mice by the injection of streptozotocin and in human renal tubular epithelial HK-2 cells cultured under a high glucose (HG, 30 mmol/L) condition. We showed that the expression levels of ACSS2 were significantly increased in renal tubular epithelial cells (RTECs) from the diabetic mice and human diabetic kidney biopsy samples, and ACSS2 was co-localized with the pro-inflammatory cytokine IL-1ß in RTECs. Diabetic ACSS2-deficient mice exhibited reduced renal tubular injury and inflammatory responses. Similarly, ACSS2 knockdown or inhibition of ACSS2 by ACSS2i (10 µmol/L) in HK-2 cells significantly ameliorated HG-induced inflammation, mitochondrial stress, and fatty acid synthesis. Molecular docking revealed that ACSS2 interacted with Sirtuin 1 (SIRT1). In HG-treated HK-2 cells, we demonstrated that ACSS2 suppressed SIRT1 expression and activated fatty acid synthesis by modulating SIRT1-carbohydrate responsive element binding protein (ChREBP) activity, leading to mitochondrial oxidative stress and inflammation. We conclude that ACSS2 promotes mitochondrial oxidative stress and renal tubular inflammation in DN by regulating the SIRT1-ChREBP pathway. This highlights the potential therapeutic value of pharmacological inhibition of ACSS2 for alleviating renal inflammation and dysregulation of fatty acid metabolic homeostasis in DN. Metabolic inflammation in the renal region, driven by lipid metabolism disorder, is a key factor in renal injury in diabetic nephropathy (DN). Acetyl-CoA synthetase 2 (ACSS2) is abundantly expressed in renal tubular epithelial cells (RTECs) and highly upregulated in diabetic kidneys. Deleting ACSS2 reduces renal fatty acid accumulation and markers of renal tubular injury in diabetic mice. We demonstrate that ACSS2 deletion inhibits ChREBP-mediated fatty acid lipogenesis, mitochondrial oxidative stress, and inflammatory response in RTECs, which play a major role in the progression of diabetic renal tubular injury in the kidney. These findings support the potential use of ACSS2 inhibitors in treating patients with DN.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Humans , Mice , Animals , Sirtuin 1/metabolism , Diabetic Nephropathies/pathology , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Molecular Docking Simulation , Kidney/pathology , Transcription Factors/metabolism , Lipid Metabolism , Glucose/metabolism , Fatty Acids/metabolism , Inflammation/metabolism , Ligases/metabolism , Lipids
4.
J Cardiovasc Pharmacol ; 82(5): 375-388, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37643027

ABSTRACT

ABSTRACT: Heart failure with preserved ejection fraction (HFpEF) is highly prevalent, accounting for 50% of all heart failure patients, and is associated with significant mortality. Sodium-glucose cotransporter subtype inhibitor (SGLT2i) is recommended in the AHA and ESC guidelines for the treatment of HFpEF, but the mechanism of SGLT2i to prevent and treat cardiac remodeling and dysfunction is currently unknown, hindering the understanding of the pathophysiology of HFpEF and the development of novel therapeutics. HFpEF model was induced by a high-fat diet (60% calories from lard) + N [w] -nitro- l -arginine methyl ester ( l -NAME-0.5 g/L) (2 Hit) in male Sprague Dawley rats to effectively recapture the myriad phenotype of HFpEF. This study's results showed that administration of dapagliflozin (DAPA, SGLT2 inhibitor) significantly limited the 2-Hit-induced cardiomyocyte hypertrophy, apoptosis, inflammation, oxidative stress, and fibrosis. It also improved cardiac diastolic and systolic dysfunction in a late-stage progression of HFpEF. Mechanistically, DAPA influences energy metabolism associated with fatty acid intake and mitochondrial dysfunction in HFpEF by increasing ß-hydroxybutyric acid (ß-OHB) levels, directing the activation of citrate synthase, reducing acetyl coenzyme A (acetyl-CoA) pools, modulating adenosine 5'-triphosphate production, and increasing the expression of mitochondrial oxidative phosphorylation system complexes I-V. In addition, following clinical DAPA therapy, the blood levels of ß-OHB and citrate synthase increased and the levels of acetyl-CoA in the blood of HFpEF patients decreased. SGLT2i plays a beneficial role in the prevention and treatment of cardiac remodeling and dysfunction in HFpEF model by attenuating cardiometabolic dysregulation.


Subject(s)
Heart Failure , Humans , Rats , Animals , Male , Heart Failure/drug therapy , Heart Failure/prevention & control , Heart Failure/metabolism , 3-Hydroxybutyric Acid/therapeutic use , Citrate (si)-Synthase , Stroke Volume/physiology , Ventricular Remodeling , Acetyl Coenzyme A/therapeutic use , Rats, Sprague-Dawley
5.
J Food Biochem ; 46(10): e14265, 2022 10.
Article in English | MEDLINE | ID: mdl-35661366

ABSTRACT

There are no medical drugs that provide an acceptable weight loss with minimal adverse effects. This study evaluated the Moringa peregrina (MP) seed extract's anti-obesity effect. Twenty-four (6/each group) male Sprague Dawley rats were divided into group Ι (control), group ΙΙ (high-fat diet [HFD]), group ΙΙΙ (HFD+ MP [250 mg/kg b.wt]), and group ΙV (HFD+ MP [500 mg/kg b.wt]). MP administration significantly ameliorated body weight gains and HFD induced elevation in cholesterol, triglycerides, LDL, and reduced HDL. Moreover, MP seed oil showed high free radical-scavenging activity, delayed ß-carotene bleaching and inhibited lipoprotein and pancreatic lipase enzymes. High-performance liquid chromatography (HPLC) revealed three major active components: crypto-chlorogenic acid, isoquercetin, and astragalin. Both quantitative Real-time PCR (RT-PCR) and western blotting revealed that MP seeds oil significantly decreased the expression of lipogenesis-associated genes such as peroxisome proliferator-activated receptors gamma (PPARγ) and fatty acid synthase (FAS) and significantly elevated the expression of lipolysis-associated genes (acetyl-CoA carboxylase1, ACCl). The oil also enhanced phosphorylation of AMP-activated protein kinase alpha (AMPK-α) and suppressed CCAAT/enhancer-binding protein ß (C/EBPß). In conclusion, administration of M. peregrina seeds oil has anti-obesity potential in HFD-induced obesity in rats. PRACTICAL APPLICATIONS: M. peregrina seeds oil had a potential anti-obesity activity that may be attributed to different mechanisms. These included decreasing body weight, and body mass index and improving lipid levels by decreasing total cholesterol, triglycerides and LDL-C, and increasing HDL-C. Also, M. peregrina seeds oil regulated adipogenesis-associated genes, such as downregulating the expression of (PPARγ, C/EBPα, and FAS) and improving and upregulating the expression and phosphorylation of AMPKα and ACCl. Despite that M. peregrina extract has reported clear anti-obesity potential through animal and laboratory studies, the available evidence-based on human clinical trials are very limited. Therefore, further studies are needed that could focus on clinical trials investigating anti-obesity potential different mechanisms of M. peregrina extract in humans.


Subject(s)
Diet, High-Fat , Moringa , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/pharmacology , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/pharmacology , Acetyl Coenzyme A/therapeutic use , Adipocytes , Animals , Antioxidants/metabolism , Body Weight , Chlorogenic Acid/metabolism , Cholesterol/metabolism , Cholesterol, LDL/metabolism , Diet, High-Fat/adverse effects , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/pharmacology , Fatty Acid Synthases/therapeutic use , Free Radicals/metabolism , Free Radicals/pharmacology , Free Radicals/therapeutic use , Humans , Lipase/metabolism , Male , Moringa/metabolism , Obesity/drug therapy , Obesity/etiology , PPAR gamma/genetics , PPAR gamma/metabolism , Plant Extracts/metabolism , Plant Extracts/pharmacology , Plant Oils/metabolism , Rats , Rats, Sprague-Dawley , Seeds/metabolism , Triglycerides/metabolism , beta Carotene
6.
Klin Lab Diagn ; (4): 27-38, 2014 Apr.
Article in Russian | MEDLINE | ID: mdl-25080798

ABSTRACT

The insulin, to provide with energy the biological function of locomotion, formed: a) pool of phylogenetically late insulin-dependent cells; b) highly productive vector variant of transfer of saturated and mono unsaturated fatty acids only to insulin-dependent cells; c) new variant of active absorption of substrates for acquiring energy by cells--apoE/B-100-receptor endocytosis; d) transformation of all endogenically synthesized palmitic saturated fatty acid in oleic mono saturated fatty acid and e) replacement of potentially ineffective palmitic variant of formation of energy in vivo with potentially high-performance oleic variant of metabolism of substrates for turning out of ATP. The insulin expressed synthesis of apoE glucose carrier 4 and stearyl-KoA-desaturase. These occurrences confirm that syndrome of insulin resistance primarily is the pathology of metabolism of fatty acids and only secondary the pathology metabolism of glucose. The multi-functional fatty cells of visceral areolar tissue and specialized adipocytes of subcutaneous fat depots are phylogenetically, regulatory and functionally different cells. They are formed under development of different biological functions: the first ones under realization of biological function of trophology and second ones under realization of biological function of locomotion. At the level of organism, the mechanisms of hypothalamus-fatty cells feedback are realized by peptide leptin and in case of hypothalamus-adipocytes feedback--peptide adiponectin. The potential possibilities of mitochondria in synthesis of ATP are high and are conditioned only by amount of substrate of mitochondria acetyl-KoA. This shortage can be chronic as in cases of disorder of insulin function and palmitic variant of metabolism of substrates for acquiring energy by cells. The deficiency of acetyl-KoA can be acute as is the case of diabetic coma when surplus amount of ketonic bodies follows the expressed deficiency of acetyl-KoA formed from glucose and fatty acids. Can the intravenous injection of acetyl-KoA be effective under diabetic ketoacidosic coma?


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Insulin/metabolism , Lipid Metabolism , Triglycerides/metabolism , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/therapeutic use , Adenosine Triphosphate/metabolism , Adipocytes , Adipose Tissue/metabolism , Biological Transport , Diabetic Coma/drug therapy , Diabetic Coma/metabolism , Humans , Hypothalamus/metabolism , Insulin Resistance , Oxidation-Reduction
7.
Nurs Times ; 110(23): 16-8, 2014.
Article in English | MEDLINE | ID: mdl-25016734

ABSTRACT

This article outlines a good practice guide to prescribing anti-dementia medication developed jointly by a lead nurse for memory services and a clinical pharmacist. The guide brings together current evidence to produce a concise prescribing guideline for practitioners.


Subject(s)
Acetyl Coenzyme A/therapeutic use , Alzheimer Disease/drug therapy , Dementia/drug therapy , Dementia/nursing , Memantine/therapeutic use , Practice Guidelines as Topic , Prescription Drugs/therapeutic use , Aged , Aged, 80 and over , Humans , Practice Patterns, Physicians' , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL
...