Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.512
Filter
1.
Nutrients ; 16(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125271

ABSTRACT

Leucine is a branched-chain amino acid that is present in protein, and it is an essential factor in activating the mechanistic target of the rapamycin complex 1 signaling pathway and increasing muscle protein synthesis. However, the loss of digestive function after total gastrectomy leads to impaired protein absorption, potentially failing to stimulate muscle protein synthesis. Therefore, this study aimed to investigate whether muscle protein synthesis is enhanced by oral skim milk administration after total gastrectomy. Male Sprague Dawley rats were divided into total gastrectomy (TG) and sham surgery (S) groups. After five weeks postoperatively, we orally administered skim milk to achieve 3.1 g protein/kg body weight and collected blood and gastrocnemius muscle. The gastrocnemius muscle weight was significantly lower in the TG group than in the S group (p < 0.05). The increase in plasma leucine concentration was significantly lower in the TG group than in the S group (p < 0.05). The skeletal muscle protein synthesis and the phosphorylation of p70S6K and 4E-BP1 showed a similar increase in both groups. Even after TG, muscle protein synthesis was stimulated by consuming skim milk, accompanied by a sufficient rise in plasma leucine concentration.


Subject(s)
Gastrectomy , Leucine , Milk , Muscle Proteins , Muscle, Skeletal , Rats, Sprague-Dawley , Animals , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle Proteins/biosynthesis , Muscle Proteins/metabolism , Leucine/administration & dosage , Leucine/pharmacology , Milk/chemistry , Phosphorylation , Rats , Administration, Oral , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Protein Biosynthesis/drug effects , Intracellular Signaling Peptides and Proteins
2.
J Med Virol ; 96(9): e29901, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39210614

ABSTRACT

The mortality and hospitalization rate by COVID-19 dropped significantly currently, but its seasonal outbreaks make antiviral treatment still vital. The mortality and hospitazation rate by COVID-19 dropped significantly currently, but its seasonal ourbreaks make antiviral treatment still vital. In our study, syrian golden hamsters were treated with molnupiravir and interferons (IFNs) after SARS-CoV-2 infection. Their weight changes, pathological changes, virus replication and inflammation levels were evaluated. In the IFNs single treatment, only IFN-α group reduced viral load (p < 0.05) and virus titer in hamster lungs. The TNF-α expression decreased significantly in both IFNs treatment at 2dpi. Histological and immunofluorescence results showed lung damage in the IFNs groups were milder at 4dpi. In the molnupiravir/IFN-α combination treatment, weight loss and virus replication in lung were significantly decreased in the mono-molnupiravir group and combination group (p < 0.05), the expression of IL-6, TNF-α, IL-1ß and MIP-1α also decreased significantly (p < 0.05), but the combination treatment was not more effective than the mono-molnupiravir treatment. Histological and immunofluorescence results showed the lung damage and inflammation in mono-molnupiravir and combination groups were milder. In summary, IFNs treatment had anti-inflammatory effect against SARS-CoV-2, only IFN-α showed a weak antiviral effect. Molnupiravir/IFN-α combination treatment was effective against SARS-CoV-2 but was not superior to mono-molnupiravir treatment. IFN-α could be considered for immunocompromised patients to stimulate and activate early immune responses.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Hydroxylamines , Lung , Mesocricetus , SARS-CoV-2 , Viral Load , Virus Replication , Animals , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Lung/virology , Lung/pathology , Lung/drug effects , Virus Replication/drug effects , SARS-CoV-2/drug effects , Viral Load/drug effects , Hydroxylamines/therapeutic use , Hydroxylamines/pharmacology , Cricetinae , Disease Models, Animal , COVID-19/immunology , COVID-19/virology , Cytidine/analogs & derivatives , Cytidine/therapeutic use , Cytidine/pharmacology , Drug Therapy, Combination , Interferon-alpha/therapeutic use , Interferon-alpha/pharmacology , Cytokines/metabolism , Interferons/therapeutic use , Male , Leucine/analogs & derivatives , Leucine/therapeutic use , Leucine/pharmacology
3.
PLoS One ; 19(8): e0309324, 2024.
Article in English | MEDLINE | ID: mdl-39163364

ABSTRACT

Plasma levels of branched-chain amino acids and their metabolites, the branched-chain ketoacids are increased in insulin resistance. Our previous studies showed that leucine and its metabolite KIC suppress insulin-stimulated glucose uptake in L6 myotubes along with the activation of the S6K1-IRS-1 pathway. Because other tissue and fiber types can be differentially regulated by KIC, we analyzed the effect of KIC gavage on whole-body insulin sensitivity and insulin signaling in vivo. We hypothesized that KIC gavage would reduce whole-body insulin sensitivity and increase S6K1-IRS-1 phosphorylation in various tissues and muscle fibers. Five-week-old male Sprague-Dawley rats were starved for 24 hours and then gavaged with 0.75ml/100g of water, leucine (22.3g/L) or KIC (30g/L) twice, ten minutes apart. They were then euthanized at different time points post-gavage (0.5-3h), and muscle, liver, and heart tissues were dissected. Other sets of gavaged animals underwent an insulin tolerance test. Phosphorylation (ph) of S6K1 (Thr389), S6 (Ser235/6) and IRS-1 (Ser612) was increased at 30 minutes post leucine gavage in skeletal muscles irrespective of fiber type. Ph-S6 (Ser235/6) was also increased in liver and heart 30 minutes after leucine gavage. KIC gavage increased ph-S6 (Ser235/6) in the liver. Neither Leucine nor KIC influenced whole-body insulin tolerance, nor ph-Akt (Ser473) in skeletal muscle and heart. BCKD-E1 α abundance was highest in the heart and liver, while ph-BCKD-E1 α (Ser293) was higher in the gastrocnemius and EDL compared to the soleus. Our data suggests that only leucine activates the S6K1-IRS-1 signaling axis in skeletal muscle, liver and heart, while KIC only does so in the liver. The effect of leucine and KIC on the S6K1-IRS-1 signaling pathway is uncoupled from whole-body insulin sensitivity. These results suggest that KIC and leucine may not induce insulin resistance, and the contributions of other tissues may regulate whole-body insulin sensitivity in response to leucine/KIC gavage.


Subject(s)
Insulin Resistance , Insulin , Keto Acids , Leucine , Rats, Sprague-Dawley , Signal Transduction , Animals , Male , Leucine/metabolism , Leucine/pharmacology , Signal Transduction/drug effects , Insulin/metabolism , Insulin/blood , Rats , Phosphorylation/drug effects , Keto Acids/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Insulin Receptor Substrate Proteins/metabolism , Liver/metabolism , Liver/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects
4.
Exp Mol Med ; 56(8): 1776-1790, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085353

ABSTRACT

Branched-chain amino acids (BCAAs), particularly leucine, are indispensable AAs for immune regulation through metabolic rewiring. However, the molecular mechanism underlying this phenomenon remains unclear. Our investigation revealed that T-cell receptor (TCR)-activated human CD4+ T cells increase the expression of BCAT1, a cytosolic enzyme responsible for BCAA catabolism, and SLC7A5, a major BCAA transporter. This upregulation facilitates increased leucine influx and catabolism, which are particularly crucial for Th17 responses. Activated CD4+ T cells induce an alternative pathway of cytosolic leucine catabolism, generating a pivotal metabolite, ß-hydroxy ß-methylbutyric acid (HMB), by acting on BCAT1 and 4-hydroxyphenylpyruvate dioxygenase (HPD)/HPD-like protein (HPDL). Inhibition of BCAT1-mediated cytosolic leucine metabolism, either with BCAT1 inhibitor 2 (Bi2) or through BCAT1, HPD, or HPDL silencing using shRNA, attenuates IL-17 production, whereas HMB supplementation abrogates this effect. Mechanistically, HMB contributes to the regulation of the mTORC1-HIF1α pathway, a major signaling pathway for IL-17 production, by increasing the mRNA expression of HIF1α. This finding was corroborated by the observation that treatment with L-ß-homoleucine (LßhL), a leucine analog and competitive inhibitor of BCAT1, decreased IL-17 production by TCR-activated CD4+ T cells. In an in vivo experimental autoimmune encephalomyelitis (EAE) model, blockade of BCAT1-mediated leucine catabolism, either through a BCAT1 inhibitor or LßhL treatment, mitigated EAE severity by decreasing HIF1α expression and IL-17 production in spinal cord mononuclear cells. Our findings elucidate the role of BCAT1-mediated cytoplasmic leucine catabolism in modulating IL-17 production via HMB-mediated regulation of mTORC1-HIF1α, providing insights into its relevance to inflammatory conditions.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Leucine , Mechanistic Target of Rapamycin Complex 1 , Signal Transduction , Th17 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Leucine/metabolism , Leucine/pharmacology , Th17 Cells/metabolism , Th17 Cells/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Humans , Animals , Mice , Cytosol/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Transaminases
5.
Fish Shellfish Immunol ; 151: 109690, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38866347

ABSTRACT

Leucine is an essential amino acid for fish. The ability of leucine to resist stress in fish has not been reported. Nitrite is a common pollutant in the aquatic environment. Therefore, we investigated the effects of dietary leucine on growth performance and nitrite-induced liver damage, mitochondrial dysfunction, autophagy, and apoptosis for sub-adult grass carp. A total of 450 grass carp (615.91 ± 1.15 g) were selected and randomly placed into 18 net cages. The leucine contents of the six diets were 2.91, 5.90, 8.92, 11.91, 14.93, and 17.92 g/kg, respectively. After a 9-week feeding trial, the nitrite exposure experiment was set up for 96 h. These results indicated that dietary leucine significantly promoted FW, WG, PWG, and SGR of sub-adult grass carp (P < 0.05). Appropriate levels of dietary leucine (11.91-17.92 g/kg) decreased the activities of serum parameters (glucose, cortisol, and methemoglobin contents, glutamic oxaloacetic transaminase, glutamic pyruvic transaminase, and lactate dehydrogenase), the contents of reactive oxygen species (ROS), nitric oxide (NO) and peroxynitrite (ONOO-). In addition, appropriate levels of dietary leucine (11.91-17.92 g/kg) increased the mRNA levels of mitochondrial biogenesis genes (PGC-1α, Nrf1/2, TFAM), fusion-related genes (Opa1, Mfn1/2) (P < 0.05), and decreased the mRNA levels of caspase 3, caspase 8, caspase 9, fission-related gene (Drp1), mitophagy-related genes (Pink1, Parkin) and autophagy-related genes (Beclin1, Ulk1, Atg5, Atg7, Atg12) (P < 0.05). Appropriate levels of dietary leucine (8.92-17.92 g/kg) also increased the protein levels of AMP-activated protein kinase (AMPK), prostacyclin (p62) and decreased the protein levels of protein light chain 3 (LC3), E3 ubiquitin ligase (Parkin), and Cytochrome c (Cytc). Appropriate levels of leucine (8.92-17.92 g/kg) could promote growth performance and alleviate nitrite-induced mitochondrial dysfunction, autophagy, apoptosis for sub-adult grass carp. Based on quadratic regression analysis of PWG and serum GPT activity, dietary leucine requirements of sub-adult grass carp were recommended to be 12.47 g/kg diet and 12.55 g/kg diet, respectively.


Subject(s)
Animal Feed , Carps , Diet , Dietary Supplements , Leucine , Nitrites , Animals , Animal Feed/analysis , Leucine/administration & dosage , Leucine/pharmacology , Diet/veterinary , Dietary Supplements/analysis , Mitochondria/drug effects , Mitochondria/metabolism , Random Allocation , Liver/drug effects , Liver/metabolism , Fish Diseases/chemically induced , Fish Diseases/prevention & control , Water Pollutants, Chemical/adverse effects , Apoptosis/drug effects , Dose-Response Relationship, Drug
6.
Diabetes ; 73(9): 1426-1439, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38870025

ABSTRACT

Glucagon is critical for the maintenance of blood glucose, however nutrient regulation of pancreatic α-cells remains poorly understood. Here, we identified a role of leucine, a well-known ß-cell fuel, in the α-cell-intrinsic regulation of glucagon release. In islet perifusion assays, physiologic concentrations of leucine strongly inhibited alanine- and arginine-stimulated glucagon secretion from human and mouse islets under hypoglycemic conditions. Mechanistically, leucine dose-dependently reduced α-cell cAMP, independently of Ca2+, ATP/ADP, or fatty acid oxidation. Leucine also reduced α-cell cAMP in islets treated with somatostatin receptor 2 antagonists or diazoxide, compounds that limit paracrine signaling from ß/δ-cells. Studies in dispersed mouse islets confirmed an α-cell-intrinsic effect. The inhibitory effect of leucine on cAMP was mimicked by glucose, α-ketoisocaproate, succinate, and the glutamate dehydrogenase activator BCH and blocked by cyanide, indicating a mechanism dependent on mitochondrial metabolism. Glucose dose-dependently reduced the impact of leucine on α-cell cAMP, indicating an overlap in function; however, leucine was still effective at suppressing glucagon secretion in the presence of elevated glucose, amino acids, and the incretin GIP. Taken together, these findings show that leucine plays an intrinsic role in limiting the α-cell secretory tone across the physiologic range of glucose levels, complementing the inhibitory paracrine actions of ß/δ-cells.


Subject(s)
Cyclic AMP , Glucagon-Secreting Cells , Glucagon , Leucine , Paracrine Communication , Animals , Glucagon/metabolism , Cyclic AMP/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/drug effects , Mice , Humans , Leucine/pharmacology , Paracrine Communication/drug effects , Glucose/metabolism , Keto Acids/pharmacology , Male , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Mice, Inbred C57BL , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism
7.
Ecotoxicol Environ Saf ; 281: 116636, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38917588

ABSTRACT

This study explored the regulatory role of bta-miR-149-3p in the inflammatory response induced by microcystin-leucine arginine (MC-LR) exposure in bovine Sertoli cells. The research endeavored to enhance the comprehension of the epigenetic mechanisms underlying MC-LR-induced cytotoxicity in Sertoli cells and establish a foundation for mitigating these effects in vitro. In this study, we elucidated the regulatory mechanism of bta-miR-149-3p in the MC-LR-induced inflammatory response by verifying the target gene of bta-miR-149-3p through luciferase assays and treating the cells with a bta-miR-149-3p inhibitor for 24 h. The results demonstrate that nuclear factor κB (NF-κB) acts as a downstream target gene of bta-miR-149-3p, which inhibits the MC-LR-induced inflammatory response in bovine Sertoli cells. This inhibition occurs by regulating the downregulation of tight junction constitutive proteins of the blood-testis barrier (BTB) through the suppression of the TLR-4/NF-κB signaling pathway (p < 0.05) and the up-regulation of the adhesion junction protein ß-catenin (p < 0.05). Notably, MC-LR exposure resulted in the up-regulation (p < 0.05) of inflammatory cytokines (IL-6, IL-1ß, and NLRP3) and the down-regulation (p < 0.05) of BTB tight junction constitutive proteins (ZO-1, Occludin) in Sertoli cells. Furthermore, the BTB constitutive protein ZO-1 exhibited significant down-regulation in Sertoli cells pretreated with the bta-miR-149-3p inhibitor compared to controls (p < 0.05), while Occludin showed no significant difference from CTNNB1 (p > 0.05). In summary, our findings suggest that bta-miR-149-3p suppresses the MC-LR-induced inflammatory response and alterations in the expression of BTB proteins in bovine Sertoli cells by inhibiting the TLR-4/NF-κB signaling pathway.


Subject(s)
Marine Toxins , MicroRNAs , Microcystins , NF-kappa B , Sertoli Cells , Signal Transduction , Toll-Like Receptor 4 , Animals , Cattle , Male , Microcystins/toxicity , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , NF-kappa B/metabolism , Signal Transduction/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Inflammation/chemically induced , Leucine/pharmacology
8.
Dent Med Probl ; 61(3): 385-390, 2024.
Article in English | MEDLINE | ID: mdl-38809132

ABSTRACT

BACKGROUND: Early colonizers adhere to the dental surface and facilitate the initial adhesion of secondary colonizers to form oral biofilms, which may cause oral infections. OBJECTIVES: This study aimed to determine the antimicrobial, anti-adhesion and antibiofilm potency of inverted amino acids on early colonizer streptococci and their mixed species. MATERIAL AND METHODS: The following test strains were used: Streptococcus gordonii (American Type Culture Collection (ATCC) 35105); Streptococcus mitis (ATCC 49456); Streptococcus oralis (ATCC 10557); Streptococcus salivarius (ATCC 7073); and Streptococcus sanguinis (ATCC BAA-1455). The concentration-dependent antimicrobial potency of d-alanine (d-ala), d-arginine (d-arg), d-leucine (d-leu), d-methionine (d-met), and d-tryptophan (d-try) was determined using the Clinical and Laboratory Standards Institute (CLSI) broth microdilution method with AlamarBlue modification. The adhesion of primary colonizers in the presence of 25-mM d-amino acids (dAAs) was assessed using the colony forming unit (CFU) assay. The CFU assay was conducted on 24-h flow cell bacterial biofilm models after exposure to 25-mM inverted dAAs. RESULTS: No minimum inhibitory concentration (MIC) point was detected at any concentration tested. The minimum bactericidal concentration (MBC) point was not observed. The adhesion of S. mitis, S. oralis and mixed species was reduced by all tested dAAs. No adverse effects were observed on S. gordonii with any of the tested dAAs. The biofilm biomass of test strains under flow conditions was significantly reduced after a 5-min exposure to all tested dAAs at 25-mM concentration. CONCLUSIONS: D-amino acids did not inhibit bacterial growth and did not show bactericidal or bacteriostatic effects on test strains at any concentration tested (ranging from 6.25 mM to 100 mM). However, dAAs effectively inhibit the adhesion of early colonizers, thereby preventing the formation of oral biofilm.


Subject(s)
Amino Acids , Bacterial Adhesion , Biofilms , Streptococcus , Biofilms/drug effects , Bacterial Adhesion/drug effects , Amino Acids/pharmacology , Amino Acids/administration & dosage , Streptococcus/drug effects , Microbial Sensitivity Tests , Humans , Biomass , Arginine/pharmacology , Streptococcus gordonii/drug effects , Anti-Bacterial Agents/pharmacology , Streptococcus oralis/drug effects , Leucine/pharmacology , Tryptophan/pharmacology
9.
J Agric Food Chem ; 72(20): 11733-11745, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38725145

ABSTRACT

Amino acids are essential for the activation of the mechanistic target of rapamycin (mTOR), but the corresponding molecular mechanism is not yet fully understood. We previously found that Met stimulated eukaryotic elongation factor α (eEF1Bα) nuclear localization in bovine mammary epithelial cells (MECs). Herein, we explored the role and molecular mechanism of eEF1Bα in methionine (Met)- and leucine (Leu)-stimulated mTOR gene transcription and milk synthesis in MECs. eEF1Bα knockdown decreased milk protein and fat synthesis, cell proliferation, and mTOR mRNA expression and phosphorylation, whereas eEF1Bα overexpression had the opposite effects. QE-MS analysis detected that eEF1Bα was phosphorylated at Ser106 in the nucleus and Met and Leu stimulated p-eEF1Bα nuclear localization. eEF1Bα knockdown abrogated the stimulation of Met and Leu by mTOR mRNA expression and phosphorylation, and this regulatory role was dependent on its phosphorylation. Akt knockdown blocked the stimulation of Met and Leu by eEF1Bα and p-eEF1Bα expression. ChIP-PCR detected that p-eEF1Bα bound only to the -548 to -793 nt site in the mTOR promoter, and ChIP-qPCR further detected that Met and Leu stimulated this binding. eEF1Bα mediated Met and Leu' stimulation on mTOR mRNA expression and phosphorylation through inducing AT-rich interaction domain 1A (ARID1A) ubiquitination degradation, and this process depended on eEF1Bα phosphorylation. p-eEF1Bα interacted with ARID1A and ubiquitin protein ligase E3 module N-recognition 5 (UBR5), and UBR5 knockdown rescued the decrease of the ARID1A protein level by eEF1Bα overexpression. Both eEF1Bα and p-eEF1Bα were highly expressed in mouse mammary gland tissues during the lactating period. In summary, we reveal that Met and Leu stimulate mTOR transcriptional activation and milk protein and fat synthesis in MECs through eEF1Bα-UBR5-ARID1A signaling.


Subject(s)
Epithelial Cells , Leucine , Methionine , Milk , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Cattle , Female , Mice , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Leucine/pharmacology , Leucine/metabolism , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/cytology , Methionine/metabolism , Methionine/pharmacology , Milk/chemistry , Milk/metabolism , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Appetite ; 200: 107509, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38795943

ABSTRACT

Glycomacropeptide (GMP) has a unique amino acid profile which may make less satiating than other dietary proteins. This study assessed the feasibility and likely acceptability of a leucine-enriched GMP drink and determined appetite response in older adults (OA). Thirteen OA (11f; 70 ± 4 years) were recruited for sensory assessments of a leucine-enriched GMP drink when mixed with water and with fruit smoothie, compared with whey protein isolate (WHEY). Participants also partook in a single focus group exploring acceptability to protein and supplementation. Separately, a counterbalanced, double-blind study with twelve OA (8f; 69 ± 3 years) was conducted to determine appetite and gut hormone responses. Fasting subjective appetite was recorded using visual analogue scales and a fasted venous blood sample was collected (to measures acyl-ghrelin, PYY, GLP-1, and CCK) before participants consumed either: GMP protein (27g + 3g leucine, 350 mL water), WHEY (30g, 350 mL water), or water. Participants rested for 240 min, with appetite measures and blood sampling throughout. An ad libitum pasta-based meal was then consumed. Sensory testing revealed low pleasantness rating for GMP in water vs. WHEY (16 ± 14 vs 31 ± 24, p = 0.016). GMP addition to smoothie reduced pleasantness (26 ± 21 vs. 61 ± 29, p = 0.009) and worsened the aroma (46 ± 15 vs. 69 ± 28, p = 0.014). The focus group revealed uncertainty of protein needs and a scepticism of supplements, with preference for food. Gut hormone response did not differ between GMP and WHEY (nAUC for all gut hormones p > 0.05). There was no difference between conditions for lunch ad libitum intake (549 ± 171 kcal, 512 ± 238 kcal, 460 ± 199 kcal for GMP, WHEY, and water, p = 0.175), or for subjective appetite response. Leucine-enriched GMP was not less satiating than WHEY, and low palatability and scepticism of supplements question the likely acceptability of GMP supplementation. Providing trusted nutritional advice and food enrichment/fortification may be preferred strategies for increasing protein intake in OA.


Subject(s)
Appetite , Caseins , Feasibility Studies , Gastrointestinal Hormones , Peptide Fragments , Whey Proteins , Humans , Female , Male , Appetite/drug effects , Aged , Pilot Projects , Gastrointestinal Hormones/blood , Double-Blind Method , Caseins/administration & dosage , Caseins/pharmacology , Whey Proteins/administration & dosage , Whey Proteins/pharmacology , Peptide Fragments/blood , Leucine/administration & dosage , Leucine/pharmacology , Ghrelin/blood , Satiation/drug effects , Eating , Dietary Supplements , Middle Aged , Peptide YY/blood , Glucagon-Like Peptide 1/blood , Dietary Proteins/administration & dosage
11.
Antiviral Res ; 227: 105904, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38729306

ABSTRACT

Despite considerable progress in developing vaccines and antivirals to combat COVID-19, the rapid mutations of the SARS-CoV-2 genome have limited the durability and efficacy of the current vaccines and therapeutic interventions. Hence, it necessitates the development of novel therapeutic approaches or repurposing existing drugs that target either viral life cycle, host factors, or both. Here, we report that SRX3177, a potent triple-activity CDK4/6-PI3K-BET inhibitor, blocks replication of the SARS-CoV-2 Omicron variant with IC50 values at sub-micromolar concentrations without any impact on the cell proliferation of Calu-3 cells at and below its IC50 concentration. When SRX3177 is combined with EIDD-1931 (active moiety of a small-molecule prodrug Molnupiravir) or MU-UNMC-2 (a SARS-CoV-2 entry inhibitor) at a fixed doses matrix, a synergistic effect was observed, leading to the significant reduction in the dose of the individual compounds to achieve similar inhibition of SARS-CoV-2 replication. Herein, we report that the combination of SRX3177/MPV or SRX3177/UM-UNMC-2 has the potential for further development as a combinational therapy against SARS-CoV-2 and in any future outbreak of beta coronavirus.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , SARS-CoV-2 , Virus Replication , SARS-CoV-2/drug effects , Humans , Antiviral Agents/pharmacology , Virus Replication/drug effects , Cytidine/analogs & derivatives , Cytidine/pharmacology , Hydroxylamines/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Virus Internalization/drug effects , Chlorocebus aethiops , Animals , Leucine/analogs & derivatives , Leucine/pharmacology , Vero Cells , Drug Synergism , Cell Line , Cyclin-Dependent Kinase 4/antagonists & inhibitors , COVID-19/virology
12.
Article in English | MEDLINE | ID: mdl-38776751

ABSTRACT

Previous research has shown that leucine (Leu) can stimulate and enhance the proliferation of equine skeletal muscle satellite cells (SCs). The gene expression profile associated with Leu-induced proliferation of equine SCs has also been documented. However, the specific role of Leu in regulating the expression of slow-twitch muscle fibers (slow-MyHC) and mitochondrial function in equine SCs, as well as the underlying mechanism, remains unclear. During this investigation, equine SCs underwent culturing in differentiation medium and were subjected to varying concentrations of Leu (0 mM, 0.5 mM, 1 mM, 2 mM, 5 mM, and 10 mM) over a span of 3 days. AMP-activated protein kinase (AMPK) inhibitor Compound C and mammalian target of rapamycin complex (mTOR) inhibitor Rapamycin were utilized to explore its underlying mechanism. Here we showed that the expression of slow-MyHC at 2 mM Leu level was significantly higher than the concentration levels of 0 mM,0.5 mM and 10 mM (P <0.01), and there was no significant difference compared to other groups (P > 0.05); the basal respiration, maximum respiration, standby respiration and the expression of slow-MyHC, PGC-1α, Cytc, ND1, TFAM, and COX1 were significantly increased with Leu supplementation (P < 0.01). We also found that Leu up-regulated the expression of key proteins on AMPK and mTOR signaling pathways, including LKB1, p-LKB1, AMPK, p-AMPK, S6, p-S6, 4EBP1, p-4EBP1, mTOR and p-mTOR (P < 0.05 or P < 0.01). Notably, when we treated the equine SCs with the AMPK inhibitor Compound C and the mTOR inhibitor Rapamycin, we observed a reduction in the beneficial effects of Leu on the expression of genes related to slow-MyHC and signaling pathway-related gene expressions. This study provides novel evidence that Leu promotes slow-MyHC expression and enhances mitochondrial function in equine SCs through the AMPK/mTOR signaling pathways, shedding light on the underlying mechanisms involved in these processes for the first time.


Subject(s)
AMP-Activated Protein Kinases , Energy Metabolism , Leucine , Muscle Fibers, Slow-Twitch , Satellite Cells, Skeletal Muscle , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Leucine/pharmacology , TOR Serine-Threonine Kinases/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/drug effects , Signal Transduction/drug effects , Horses , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Energy Metabolism/drug effects , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/drug effects , Cells, Cultured
13.
Cancer Sci ; 115(7): 2461-2472, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38655663

ABSTRACT

L-type amino acid transporter 1 (LAT1) is specifically expressed in many malignancies, contributes to the transport of essential amino acids, such as leucine, and regulates the mammalian target of rapamycin (mTOR) signaling pathway. We investigated the expression profile and functional role of LAT1 in prostate cancer using JPH203, a specific inhibitor of LAT1. LAT1 was highly expressed in castration-resistant prostate cancer (CRPC) cells, including C4-2 and PC-3 cells, but its expression level was low in castration-sensitive LNCaP cells. JPH203 significantly inhibited [14C] leucine uptake in CRPC cells but had no effect in LNCaP cells. JPH203 inhibited the proliferation, migration, and invasion of CRPC cells but not of LNCaP cells. In C4-2 cells, Cluster of differentiation (CD) 24 was identified by RNA sequencing as a novel downstream target of JPH203. CD24 was downregulated in a JPH203 concentration-dependent manner and suppressed activation of the Wnt/ß-catenin signaling pathway. Furthermore, an in vivo study showed that JPH203 inhibited the proliferation of C4-2 cells in a castration environment. The results of this study indicate that JPH203 may exert its antitumor effect in CRPC cells via mTOR and CD24.


Subject(s)
CD24 Antigen , Cell Movement , Cell Proliferation , Large Neutral Amino Acid-Transporter 1 , Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Large Neutral Amino Acid-Transporter 1/metabolism , Cell Line, Tumor , Animals , Cell Proliferation/drug effects , CD24 Antigen/metabolism , Mice , Cell Movement/drug effects , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Benzoxazoles/pharmacology , Leucine/pharmacology , Leucine/analogs & derivatives , Mice, Nude , Gene Expression Regulation, Neoplastic/drug effects , Tyrosine/analogs & derivatives
14.
Metabolism ; 156: 155920, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38677663

ABSTRACT

BACKGROUND: Statins, or hydroxy-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors, are one of the most commonly prescribed medications for lowering cholesterol. Myopathic side-effects ranging from pain and soreness to critical rhabdomyolysis are commonly reported and often lead to discontinuation. The pathophysiological mechanism is, in general, ascribed to a downstream reduction of Coenzyme Q10 synthesis. HMG-CoA is a metabolite of leucine and its corresponding keto acid α-ketoisocaproic acid (KIC) and ß-hydroxy-ß-methylbutyrate (HMB), however, little is known about the changes in the metabolism of leucine and its metabolites in response to statins. OBJECTIVE: We aimed to investigate if statin treatment has implications on the upstream metabolism of leucine to KIC and HMB, as well as on other branched chain amino acids (BCAA). DESIGN: 12 hyperlipidemic older adults under statin treatment were recruited. The study was conducted as a paired prospective study. Included participants discontinued their statin treatment for 4 weeks before they returned for baseline measurements (before). Statin treatment was then reintroduced, and the participants returned for a second study day 7 days after reintroduction (after statin). On study days, participants were injected with stable isotope pulses for measurement of the whole-body production (WBP) of all BCAA (leucine, isoleucine and valine), along with their respective keto acids and HMB. RESULTS: We found a reduced leucine WBP (22 %, p = 0.0033), along with a reduction in valine WBP (13 %, p = 0.0224). All other WBP of BCAA and keto acids were unchanged. There were no changes in the WBP of HMB. CONCLUSIONS: Our study shows that statin inhibition of HMG-CoA reductase has an upstream impact on the turnover of leucine and valine. Whether this impairment in WBP of leucine may contribute to the known pathophysiological side effects of statins on muscle remains to be further investigated.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Leucine , Valerates , Leucine/metabolism , Leucine/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Humans , Valerates/pharmacology , Male , Female , Aged , Prospective Studies , Middle Aged , Hyperlipidemias/drug therapy , Hyperlipidemias/metabolism , Keto Acids/metabolism , Amino Acids, Branched-Chain/metabolism
15.
J Sci Food Agric ; 104(11): 6696-6705, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38551359

ABSTRACT

BACKGROUND: Leucine (Leu) is an essential amino acid that facilitates skeletal muscle satellite cell differentiation, yet its mechanism remains underexplored. Sestrin2 (SESN2) serves as a Leu sensor, binding directly to Leu, while ribophorin II (RPN2) acts as a signaling factor in multiple pathways. This study aimed to elucidate Leu's impact on mouse C2C12 cell differentiation and skeletal muscle injury repair by modulating RPN2 expression through SESN2, offering a theoretical foundation for clinical skeletal muscle injury prevention and treatment. RESULTS: Leu addition promoted C2C12 cell differentiation compared to the control, enhancing early differentiation via myogenic determinant (MYOD) up-regulation. Sequencing revealed SESN2 binding to and interacting with RPN2. RPN2 overexpression up-regulated MYOD, myogenin and myosin heavy chain 2, concurrently decreased p-GSK3ß and increased nuclear ß-catenin. Conversely, RPN2 knockdown yielded opposite results. Combining RPN2 knockdown with Leu rescued increased p-GSK3ß and decreased nuclear ß-catenin compared to Leu absence. Hematoxylin and eosin staining results showed that Leu addition accelerated mouse muscle damage repair, up-regulating Pax7, MYOD and RPN2 in the cytoplasm, and nuclear ß-catenin, confirming that the role of Leu in muscle injury repair was consistent with the results for C2C12 cells. CONCLUSION: Leu, bound with SESN2, up-regulated RPN2 expression, activated the GSK3ß/ß-catenin pathway, enhanced C2C12 differentiation and expedited skeletal muscle damage repair. © 2024 Society of Chemical Industry.


Subject(s)
Cell Differentiation , Glycogen Synthase Kinase 3 beta , Leucine , Signal Transduction , beta Catenin , Animals , Mice , beta Catenin/metabolism , beta Catenin/genetics , Cell Line , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Leucine/metabolism , Leucine/pharmacology , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Myoblasts/metabolism , Myoblasts/cytology , MyoD Protein/metabolism , MyoD Protein/genetics , Myogenin/metabolism , Myogenin/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Sestrins
16.
Elife ; 122024 Mar 05.
Article in English | MEDLINE | ID: mdl-38442142

ABSTRACT

Cytokine storms are associated with severe pathological damage and death in some diseases. Excessive activation of M1 macrophages and the subsequent secretion of pro-inflammatory cytokines are a major cause of cytokine storms. Therefore, promoting the polarization of M2 macrophages to restore immune balance is a promising therapeutic strategy for treating cytokine storm syndrome (CSS). This study was aimed at investigating the potential protective effects of leucine on lipopolysaccharide (LPS)-induced CSS in mice and exploring the underlying mechanisms. CSS was induced by LPS administration in mice, which were concurrently administered leucine orally. In vitro, bone marrow derived macrophages (BMDMs) were polarized to M1 and M2 phenotypes with LPS and interleukin-4 (IL-4), respectively, and treated with leucine. Leucine decreased mortality in mice treated with lethal doses of LPS. Specifically, leucine decreased M1 polarization and promoted M2 polarization, thus diminishing pro-inflammatory cytokine levels and ameliorating CSS in mice. Further studies revealed that leucine-induced macrophage polarization through the mechanistic target of rapamycin complex 1 (mTORC1)/liver X receptor α (LXRα) pathway, which synergistically enhanced the expression of the IL-4-induced M2 marker Arg1 and subsequent M2 polarization. In summary, this study revealed that leucine ameliorates CSS in LPS mice by promoting M2 polarization through the mTORC1/LXRα/Arg1 signaling pathway. Our findings indicate that a fundamental link between metabolism and immunity contributes to the resolution of inflammation and the repair of damaged tissues.


Subject(s)
Cytokine Release Syndrome , Interleukin-4 , Animals , Mice , Liver X Receptors , Leucine/pharmacology , Lipopolysaccharides , Cytokines , Signal Transduction , Macrophages , Mechanistic Target of Rapamycin Complex 1
17.
Molecules ; 29(5)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38474693

ABSTRACT

Antimicrobial peptides (AMPs), acknowledged as host defense peptides, constitute a category of predominant cationic peptides prevalent in diverse life forms. This study explored the antibacterial activity of α-conotoxin RgIA, and to enhance its stability and efficacy, D-amino acid substitution was employed, resulting in the synthesis of nine RgIA mutant analogs. Results revealed that several modified RgIA mutants displayed inhibitory efficacy against various pathogenic bacteria and fungi, including Candida tropicalis and Escherichia coli. Mechanistic investigations elucidated that these polypeptides achieved antibacterial effects through the disruption of bacterial cell membranes. The study further assessed the designed peptides' hemolytic activity, cytotoxicity, and safety. Mutants with antibacterial activity exhibited lower hemolytic activity and cytotoxicity, with Pep 8 demonstrating favorable safety in mice. RgIA mutants incorporating D-amino acids exhibited notable stability and adaptability, sustaining antibacterial properties across diverse environmental conditions. This research underscores the potential of the peptide to advance innovative oral antibiotics, offering a novel approach to address bacterial infections.


Subject(s)
Anti-Infective Agents , Conotoxins , Mice , Animals , Lysine/pharmacology , Leucine/pharmacology , Amino Acid Substitution , Conotoxins/chemistry , Anti-Infective Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacteria , Microbial Sensitivity Tests
18.
Biomed Pharmacother ; 174: 116480, 2024 May.
Article in English | MEDLINE | ID: mdl-38547765

ABSTRACT

Sepsis is caused by an inadequate or dysregulated host response to infection. Enzymes causing cellular degradation are matrix metalloproteinases (MMPs). Lipopolysaccharide (LPS) is used in models of sepsis in laboratory settings The aim of the study was to measure MMP 2 and 12 concentrations in spleen and lungs in rats in which septic shock was induced by LPS. The experiment was carried out on 40 male Wistar rats (5 groups of 8): 0. controls 1. administered LPS 2. administered bestatin 3. LPS and bestatin 4.bestatin and after 6 hours LPS Animals were decapitated. Lungs and spleens were collected. Concentrations of MMP-2 and MMP-12 were determined using immunoenzymatic methods. Mean (±SD) MMP-2 in the controls was 43.57 ± 20.53 ng/ml in the lungs and 1.7 ± 0.72 ng/ml in the spleen; Group 1: 31.28 ± 13.13 ng/ml, 0.83 ± 0.8 ng/ml; Group 2: 44.24 ± 22.75 ng /ml, 1.01 ± 0.32 ng/ml; Group 3: 35.94 ± 15.13 ng/ml, 0.41 ± 0.03 ng/ml; Group 4:79.42 ± 44.70 ng/ml, 0.45 ± 0.15, respectively. Mean MMP-12 in controls was 19.79 ± 10.01 ng/ml in lungs and 41.13 ± 15.99 ng/ml in the spleen; Group 1:27.97 ± 15.1 ng/ml; 40.44 ± 11.2 ng/ml; Group 2: 37.93 ± 25.38 ng/ml 41.05 ± 18.08 ng/ml; Group 3: 40.59 ± 11.46 ng/ml, 35.16 ± 12.89 ng/ml; Group 4: 39.4 ± 17.83 ng/ml, 42.04 ± 12.35 ng/ml, respectively. CONCLUSIONS: 1. Bestatin reduces MMP 2 and 12 levels in spleen and lungs. 2. Treatment with bestatin minimizes the effect of LPS.


Subject(s)
Disease Models, Animal , Leucine , Leucine/analogs & derivatives , Lipopolysaccharides , Lung , Matrix Metalloproteinase 12 , Matrix Metalloproteinase 2 , Rats, Wistar , Sepsis , Spleen , Animals , Spleen/drug effects , Spleen/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Lung/drug effects , Lung/enzymology , Lung/pathology , Lung/metabolism , Sepsis/drug therapy , Sepsis/chemically induced , Matrix Metalloproteinase 12/metabolism , Rats , Leucine/pharmacology , Leucine/therapeutic use , Matrix Metalloproteinase Inhibitors/pharmacology
19.
Nat Metab ; 6(2): 359-377, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38409323

ABSTRACT

High protein intake is common in western societies and is often promoted as part of a healthy lifestyle; however, amino-acid-mediated mammalian target of rapamycin (mTOR) signalling in macrophages has been implicated in the pathogenesis of ischaemic cardiovascular disease. In a series of clinical studies on male and female participants ( NCT03946774 and NCT03994367 ) that involved graded amounts of protein ingestion together with detailed plasma amino acid analysis and human monocyte/macrophage experiments, we identify leucine as the key activator of mTOR signalling in macrophages. We describe a threshold effect of high protein intake and circulating leucine on monocytes/macrophages wherein only protein in excess of ∼25 g per meal induces mTOR activation and functional effects. By designing specific diets modified in protein and leucine content representative of the intake in the general population, we confirm this threshold effect in mouse models and find ingestion of protein in excess of ∼22% of dietary energy requirements drives atherosclerosis in male mice. These data demonstrate a mechanistic basis for the adverse impact of excessive dietary protein on cardiovascular risk.


Subject(s)
Cardiovascular Diseases , Humans , Male , Female , Mice , Animals , Leucine/metabolism , Leucine/pharmacology , Risk Factors , TOR Serine-Threonine Kinases/metabolism , Macrophages/metabolism , Heart Disease Risk Factors , Mammals/metabolism
20.
Poult Sci ; 103(4): 103509, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38387289

ABSTRACT

Light pollution is a potential risk factor for intestinal health. Tryptophan plays an important role in the inhibition of intestinal inflammation. However, the mechanism of tryptophan in alleviating intestinal inflammation caused by long photoperiod is still unclear. This study investigated the anti-inflammatory effect of dietary tryptophan on intestinal inflammatory damage induced by long photoperiod and its potential mechanism in broiler chickens. We found that dietary tryptophan mitigated long photoperiod-induced intestinal tissue inflammatory damage and inhibited the activation of Nucleotide-Binding Oligomerization Domain, Leucine-Rich Repeat and Pyrin Domain-Containing 3 inflammasome. Moreover, dietary tryptophan significantly increased the relative abundance of Faecalibacterium, Enterococcus, and Lachnospiraceae_NC2004_group were significantly decreased the relative abundance of Ruminococcus_torques_group and norank_f_UCG-010 under the condition of long photoperiod (P < 0.05). The results of tryptophan targeted metabolomics show that tryptophan significantly increased indole-3-acetic acid (IAA) and indole-3 lactic acid (ILA), and significantly decreased xanthurenic acid (XA) under long photoperiod (P < 0.05). In conclusion, the results indicated that dietary tryptophan alleviates intestinal inflammatory damage caused by long photoperiod via the inhibition of Nucleotide-Binding Oligomerization Domain, Leucine-Rich Repeat and Pyrin Domain-Containing 3 inflammasome activation, which was mediated by tryptophan metabolites. Therefore, tryptophan supplementation could be a promising way to protect the intestine health under the condition of long photoperiod.


Subject(s)
Gastrointestinal Microbiome , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Chickens/physiology , Tryptophan/pharmacology , Tryptophan/metabolism , Leucine/pharmacology , Photoperiod , Inflammation/veterinary , Nucleotides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL