High-salt diets (HSDs) are known to impact blood pressure and cardiovascular health, but their effects on glucose metabolism, liver function, and gut microbiota remain poorly understood. This study investigates how long-term HSD affects these physiological processes and evaluates the potential therapeutic effects of ACE inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs). Male Sprague-Dawley rats were fed a normal salt diet (0.3% NaCl), a moderate salt diet (2% NaCl), or a high-salt diet (8% NaCl) for 12 wk. Two subgroups in the HSD condition received telmisartan or enalapril. We assessed blood pressure, glucose homeostasis, liver inflammation, pancreatic function, and gut microbiota composition. HSD rats exhibited significantly higher blood pressure [130 ± 2 mmHg in normal diet (ND) vs. 144 ± 4 mmHg in HSD; P < 0.01], reduced fasting insulin (1.33 ± 0.14 ng/mL in ND vs. 0.60 ± 0.05 ng/mL in HSD; P < 0.01), and gut microbiota dysbiosis, with a 71% reduction in Ruminococcus species (P = 0.018). Liver inflammation, indicated by an increase in CD68+ macrophages, was also observed in the HSD group. Telmisartan treatment significantly reduced liver inflammation but did not fully restore metabolic homeostasis. HSD disrupts multiple physiological systems, including glucose metabolism and liver function, partly through gut microbiota alterations. ACEIs and ARBs provided partial protection, highlighting the need for multitargeted interventions to mitigate high-salt diet effects.NEW & NOTEWORTHY High-salt diet induces multisystem disruptions, including liver inflammation, reduced insulin levels, and gut microbiota imbalance. ACEIs and ARBs showed limited efficacy, highlighting the need for comprehensive therapeutic approaches.
- MeSH
- Angiotensin Receptor Antagonists * pharmacology MeSH
- Enalapril pharmacology MeSH
- Glucose * metabolism MeSH
- Angiotensin-Converting Enzyme Inhibitors * pharmacology MeSH
- Liver * drug effects metabolism MeSH
- Blood Glucose metabolism drug effects MeSH
- Blood Pressure drug effects MeSH
- Rats MeSH
- Sodium Chloride, Dietary * adverse effects MeSH
- Rats, Sprague-Dawley MeSH
- Gastrointestinal Microbiome * drug effects MeSH
- Telmisartan pharmacology MeSH
- Animals MeSH
- Check Tag
- Rats MeSH
- Male MeSH
- Animals MeSH
- Publication type
- Journal Article MeSH
Kontext: Infekční onemocnění covid-19 je spojeno s endotelovou dysfunkcí. Ověřuje se představa možnosti opravy endotelové buňky pomocí endotelových progenitorových buněk (endothelial progenitor cell, EPC) s použitím inhibitorů angiotenzin konvertujícího enzymu (ACEI); podle této představy by se tak mohla zlepšit angiogeneze EPC. Metody: Jedná se o skutečně experimentální in vitro studii s uspořádáním s kontrolní skupinou vytvořenou až po testu. Mononukleární buňky byly izolovány z periferní žilní krve pacienta s chronickým koronárním syndromem, který měl v anamnéze infekční onemocnění covid-19. Buňky byly kultivovány ve speciálním médiu po dobu sedmi dní a buňky EPC byly identifikovány imunocytochemicky pomocí protilátek proti buňkám CD34 radioaktivně značeným fluoresceinizothiocyanátem (FITC) s vyšetřením pod fluorescenčním mikroskopem. Buňky byly rozděleny do dvou skupin, kontrolní a do skupiny s aplikací lisinoprilu v dávce 50 μM. Morfologie a spojení tubulů byly zkoumány pomocí analyzátoru obrázků výrobce Wimasis. Koncentrace von Willebrandova faktoru (vWF) a CD31 byly měřeny metodou ELISA, následovanou statistickým srovnáním obou skupin, přičemž za statisticky významnou byla považována hodnota p < 0,05. Výsledky: Po 144hodinové kultivaci byly buňky EPC identifikovány pomocí světelné a fluorescenční mikroskopie. Parametry tvorby tubulů včetně pokryté plochy (29,6 ± 15,68 vs. 61,8 ± 25,41; p 0,13), celkového počtu tubulů 387 ± 101,55 vs. 382,67 ± 158,53; p 0,97), větvení (163 ± 72,52 vs. 179,66 ± 53,5; p 0,543) a celkového počtu kliček (40,66 ± 30,73 vs. 52,66 ± 5,77; p 0,543) nevykazovaly mezi kontrolní skupinou a skupinou s aplikací lisinoprilu žádný rozdíl. Pokud se týče sérologických biomarkerů, hodnoty CD31 se mezi kontrolami a skupinou s aplikací lisinoprilu statisticky významně nelišily (2 903,58 ± 578,08 vs. 3 361,89 ± 391,24; p 0,319), nicméně hodnoty vWF byly statisticky významně vyšší ve skupině s aplikací lisinoprilu (98,670 ± 3,240 vs. 91,181 ± 2,443; p 0,033). Závěr: U pacientů se stabilní ischemickou chorobou srdeční po prodělaném infekčním onemocnění covid-19 může lisinopril ovlivňovat angiogenezi buněk EPC, jak dokazuje zvyšování hodnot parametrů tvorby tubulů a statisticky významný nárůst koncentrace von Willebrandova faktoru.
Background: COVID-19 infection is associated with endothelial dysfunction. The concept of endothelial cell repair utilizing endothelial progenitor cells (EPCs) with the use of angiotensin-converting enzyme inhibitors (ACEIs) has been developing which is known for its potential for EPC's angiogenesis improvement. Methods: This is a true experimental in vitro study with post-test only control group design. Mononuclear cells were isolated from peripheral venous blood of patient with chronic coronary syndrome and history of COVID-19. The cells then cultured on special media for 7 days and EPC was identified using immunocytochemical examination with labelled anti-CD34 cells FITC-under fluorescence microscope examination. The cells were divided into two groups consisted of control group and 50 μM lisinopril-treated group. The morphology and tube connections were analyzed using Wimasis image analyzer. The von Willebrand factor (vWF) and CD31 concentrations were also measured by ELISA. Statistical comparison between both groups was performed and p-value < 0.05 was considered significant.
- MeSH
- Angiogenesis drug effects MeSH
- Antigens, CD34 MeSH
- COVID-19 complications MeSH
- Adult MeSH
- Endothelial Progenitor Cells * drug effects MeSH
- Microscopy, Fluorescence MeSH
- Immunohistochemistry methods instrumentation MeSH
- Coronary Disease drug therapy complications MeSH
- Humans MeSH
- Lisinopril * administration & dosage MeSH
- Statistics as Topic MeSH
- In Vitro Techniques MeSH
- Check Tag
- Adult MeSH
- Humans MeSH
- Male MeSH
- Publication type
- Clinical Study MeSH
- Research Support, Non-U.S. Gov't MeSH
BACKGROUND: [177Lu]Lu-PSMA-617 (177Lu-PSMA-617) prolongs radiographic progression-free survival and overall survival in patients with metastatic castration-resistant prostate cancer previously treated with androgen receptor pathway inhibitor (ARPI) and taxane therapy. We aimed to investigate the efficacy of 177Lu-PSMA-617 in patients with taxane-naive metastatic castration-resistant prostate cancer. METHODS: In this phase 3, randomised, controlled trial conducted at 74 sites across Europe and North America, taxane-naive patients with prostate-specific membrane antigen (PSMA)-positive metastatic castration-resistant prostate cancer who had progressed once on a previous ARPI were randomly allocated (1:1) to open-label, intravenous 177Lu-PSMA-617 at a dosage of 7·4 GBq (200 mCi) ± 10% once every 6 weeks for six cycles, or a change of ARPI (to abiraterone or enzalutamide, administered orally on a continuous basis per product labelling). Crossover from ARPI change to 177Lu-PSMA-617 was allowed after centrally confirmed radiographic progression. The primary endpoint was radiographic progression-free survival, defined as the time from randomisation until radiographic progression or death, assessed in the intention-to-treat population. Safety was a secondary endpoint. This study is registered with ClinicalTrials.gov (NCT04689828) and is ongoing. In this primary report of the study, we present primary (first data cutoff) and updated (third data cutoff) analyses of radiographic progression-free survival; all other data are based on the third data cutoff. FINDINGS: Overall, of the 585 patients screened, 468 met all eligibility criteria and were randomly allocated between June 15, 2021 and Oct 7, 2022 to receive 177Lu-PSMA-617 (234 [50%] patients) or ARPI change (234 [50%]). Baseline characteristics were mostly similar between groups; median number of 177Lu-PSMA-617 cycles was 6·0 (IQR 4·0-6·0). Of patients assigned to ARPI change, 134 (57%) crossed over to receive 177Lu-PSMA-617. In the primary analysis (median time from randomisation to first data cutoff 7·26 months [IQR 3·38-10·55]), the median radiographic progression-free survival was 9·30 months (95% CI 6·77-not estimable) in the 177Lu-PSMA-617 group versus 5·55 months (4·04-5·95) in the ARPI change group (hazard ratio [HR] 0·41 [95% CI 0·29-0·56]; p<0·0001). In the updated analysis at time of the third data cutoff (median time from randomisation to third data cutoff 24·11 months [IQR 20·24-27·40]), median radiographic progression-free survival was 11·60 months (95% CI 9·30-14·19) in the 177Lu-PSMA-617 group versus 5·59 months (4·21-5·95) in the ARPI change group (HR 0·49 [95% CI 0·39-0·61]). The incidence of grade 3-5 adverse events was lower in the 177Lu-PSMA-617 group (at least one event in 81 [36%] of 227 patients; four [2%] grade 5 [none treatment related]) than the ARPI change group (112 [48%] of 232; five [2%] grade 5 [one treatment related]). INTERPRETATION: 177Lu-PSMA-617 prolonged radiographic progression-free survival relative to ARPI change, with a favourable safety profile. For patients with PSMA-positive metastatic castration-resistant prostate cancer who are being considered for a change of ARPI after progression on a previous ARPI, 177Lu-PSMA-617 may be an effective treatment alternative. FUNDING: Novartis.
- MeSH
- Androstenes * therapeutic use MeSH
- Androgen Receptor Antagonists therapeutic use MeSH
- Benzamides therapeutic use MeSH
- Dipeptides * therapeutic use MeSH
- Progression-Free Survival MeSH
- Phenylthiohydantoin * therapeutic use MeSH
- Heterocyclic Compounds, 1-Ring * therapeutic use MeSH
- Middle Aged MeSH
- Humans MeSH
- Lutetium * therapeutic use MeSH
- Prostatic Neoplasms, Castration-Resistant * drug therapy pathology MeSH
- Nitriles * therapeutic use MeSH
- Prostate-Specific Antigen blood MeSH
- Radioisotopes therapeutic use MeSH
- Aged MeSH
- Taxoids therapeutic use MeSH
- Check Tag
- Middle Aged MeSH
- Humans MeSH
- Male MeSH
- Aged MeSH
- Publication type
- Journal Article MeSH
- Clinical Trial, Phase III MeSH
- Multicenter Study MeSH
- Randomized Controlled Trial MeSH
Thank you very much for your comment [...].
- MeSH
- beta-Alanine * administration & dosage MeSH
- Carnosine * MeSH
- Humans MeSH
- Dietary Supplements * MeSH
- Check Tag
- Humans MeSH
- Publication type
- Journal Article MeSH
Nephrotoxicity as a cause of acute kidney injury (AKI) induced by cisplatin (CP), limits its usefulness as an anticancer agent. Diminazene, an angiotensin converting enzyme 2 activator, exhibited renoprotective properties on rat models of kidney diseases. This research aims to investigate the salutary effect of diminazene in comparison with lisinopril or valsartan in CP-induced AKI. The first and second groups of rats received oral vehicle (distilled water) for 9 days, and saline injection or intraperitoneal CP (6 mg/kg) on day 6, respectively. Third, fourth, and fifth groups received intraperitoneal injections of CP on day 6 and diminazene (15 mg/kg/day, orally), lisinopril (10 mg/kg/day, orally), or valsartan (30 mg/kg/day, orally), for 9 days, respectively. 24h after the last day of treatment, blood and kidneys were removed under anesthesia for biochemical and histopathological examination. Urine during the last 24 h before sacrificing the rats was also collected. CP significantly increased plasma urea, creatinine, neutrophil gelatinase-associated lipocalin, calcium, phosphorus, and uric acid. It also increased urinary albumin/creatinine ratio, N-Acetyl-beta-D-Glucosaminidase/creatinine ratio, and reduced creatinine clearance, as well the plasma concentrations of inflammatory cytokines [plasma tumor necrosis factor-alpha, and interleukin-1beta], and significantly reduced antioxidant indices [catalase, glutathione reductase , and superoxide dismutase]. Histopathologically, CP treatment caused necrosis of renal tubules, tubular casts, shrunken glomeruli, and increased renal fibrosis. Diminazine, lisinopril, and valsartan ameliorated CP-induced biochemical and histopathological changes to a similar extent. The salutary effect of the three drugs used is, at least partially, due to their anti-inflammatory and antioxidant effects. Keywords: Cisplatin, Diminazene, ACE2 activator, Lisinopril, Valsartan, Acute kidney injury.
- MeSH
- Acute Kidney Injury * chemically induced pathology metabolism prevention & control drug therapy MeSH
- Cisplatin * toxicity MeSH
- Diminazene * analogs & derivatives pharmacology therapeutic use MeSH
- Angiotensin-Converting Enzyme Inhibitors pharmacology MeSH
- Rats MeSH
- Kidney drug effects pathology metabolism MeSH
- Lisinopril * pharmacology MeSH
- Rats, Wistar * MeSH
- Antineoplastic Agents toxicity MeSH
- Valsartan * pharmacology MeSH
- Animals MeSH
- Check Tag
- Rats MeSH
- Male MeSH
- Animals MeSH
- Publication type
- Journal Article MeSH
- Comparative Study MeSH
Glutamate carboxypeptidase II (GCPII, also known as PSMA or FOLH1) is responsible for the cleavage of N-acetyl-aspartyl-glutamate (NAAG) to N-acetyl-aspartate and glutamate in the central nervous system and facilitates the intestinal absorption of folate by processing dietary folyl-poly-γ-glutamate in the small intestine. The physiological function of GCPII in other organs like kidneys is still not known. GCPII inhibitors are neuroprotective in various conditions (e.g., ischemic brain injury) in vivo; however, their utilization as potential drug candidates has not been investigated in regard to not yet known GCPII activities. To explore the GCPII role and possible side effects of GCPII inhibitors, we performed parallel metabolomic and lipidomic analysis of the cerebrospinal fluid (CSF), urine, plasma, and brain tissue of mice with varying degrees of GCPII deficiency (fully deficient in Folh1, -/-; one allele deficient in Folh1, +/-; and wild type, +/+). Multivariate analysis of metabolites showed no significant differences between wild-type and GCPII-deficient mice (except for NAAG), although changes were observed between the sex and age. NAAG levels were statistically significantly increased in the CSF, urine, and plasma of GCPII-deficient mice. However, no difference in NAAG concentrations was found in the whole brain lysate likely because GCPII, as an extracellular enzyme, can affect only extracellular and not intracellular NAAG concentrations. Regarding the lipidome, the most pronounced genotype-linked changes were found in the brain tissue. In brains of GCPII-deficient mice, we observed statistically significant enrichment in phosphatidylcholine-based lipids and reduction of sphingolipids and phosphatidylethanolamine plasmalogens. We hypothesize that the alteration of the NAA-NAAG axis by absent GCPII activity affected myelin composition. In summary, the absence of GCPII and thus similarly its inhibition do not have detrimental effects on metabolism, with just minor changes in the brain lipidome.
- MeSH
- Dipeptides metabolism MeSH
- Glutamate Carboxypeptidase II * genetics metabolism MeSH
- Glutamic Acid MeSH
- Lipidomics * MeSH
- Lipids chemistry MeSH
- Metabolomics * MeSH
- Brain metabolism MeSH
- Mice MeSH
- Animals MeSH
- Check Tag
- Mice MeSH
- Animals MeSH
- Publication type
- Journal Article MeSH
- Research Support, Non-U.S. Gov't MeSH
Carnosine is a performance-enhancing food supplement with a potential to modulate muscle energy metabolism and toxic metabolites disposal. In this study we explored interrelations between carnosine supplementation (2 g/day, 12 weeks) induced effects on carnosine muscle loading and parallel changes in (i) muscle energy metabolism, (ii) serum albumin glycation and (iii) reactive carbonyl species sequestering in twelve (M/F=10/2) sedentary, overweight-to-obese (BMI: 30.0+/-2.7 kg/m2) adults (40.1+/-6.2 years). Muscle carnosine concentration (Proton Magnetic Resonance Spectroscopy; 1H-MRS), dynamics of muscle energy metabolism (Phosphorus Magnetic Resonance Spectroscopy; 31P-MRS), body composition (Magnetic Resonance Imaging; MRI), resting energy expenditure (indirect calorimetry), glucose tolerance (oGTT), habitual physical activity (accelerometers), serum carnosine and carnosinase-1 content/activity (ELISA), albumin glycation, urinary carnosine and carnosine-propanal concentration (mass spectrometry) were measured. Supplementation-induced increase in muscle carnosine was paralleled by improved dynamics of muscle post-exercise phosphocreatine recovery, decreased serum albumin glycation and enhanced urinary carnosine-propanal excretion (all p<0.05). Magnitude of supplementation-induced muscle carnosine accumulation was higher in individuals with lower baseline muscle carnosine, who had lower BMI, higher physical activity level, lower resting intramuscular pH, but similar muscle mass and dietary protein preference. Level of supplementation-induced increase in muscle carnosine correlated with reduction of protein glycation, increase in reactive carbonyl species sequestering, and acceleration of muscle post-exercise phosphocreatine recovery.
- MeSH
- Adult MeSH
- Phosphocreatine metabolism MeSH
- Carnosine * metabolism pharmacology MeSH
- Muscle, Skeletal metabolism MeSH
- Humans MeSH
- Maillard Reaction MeSH
- Dietary Supplements MeSH
- Check Tag
- Adult MeSH
- Humans MeSH
- Publication type
- Journal Article MeSH
Biomonitoring of human exposure to reactive electrophilic chemicals such as ethylene oxide (EO) has been commonly based on the determination of adducts with N-terminal valine in blood protein globin, but a systematic search has also been undertaken to find surrogate markers enabling non-invasive sampling. Recently, N-(2-hydroxyethyl)-L-valyl-L-leucine (HEVL) has been identified as an ultimate cleavage product of EO-adducted globin in the urine of occupationally exposed workers. Herein, full validation of the analytical procedure consisting of solid-phase extraction of HEVL from urine samples (2 mL) followed by high-performance liquid chromatography-electrospray ionization-high-resolution mass spectrometry determination using deuterium-labeled HEVL as an internal standard (IS) is described. Method limit of quantitation is 0.25 ng/mL, and its selectivity is excellent as demonstrated by the invariable ratio of the qualifier and quantifier ion intensities across diverse urine samples and synthetic standard. The linear calibration model was applicable over the whole concentration range tested (0.25-10 ng/mL). The method accuracy assessed as a recovery of HEVL using a spiking experiment was 98-100%. Within-day precision of the method ranged from 1.8% to 3.0%, while the results from consecutive analytical runs conducted within 1 week or within 10-150 weeks differed in the range of 2.2-9.7%. The stability study on urine samples (-20°C up to 3 years, freeze-and-thaw up to 10 cycles) as well as on aqueous solutions (5°C up to 4 months) indicated no relevant changes in HEVL concentration (≤4%) over the time tested. Analytical responses of both HEVL and IS correlated with urinary creatinine as an index of matrix composition, but this matrix effect was mostly eliminated using the HEVL/IS peak area ratio, attaining the IS-normalized relative matrix effect <3%. In conclusion, the method complied successfully with the bioanalytical method validation criteria, making it a reliable tool for HEVL determination in human biomonitoring.
- MeSH
- Dipeptides * MeSH
- Ethylene Oxide * MeSH
- Globins MeSH
- Leucine MeSH
- Humans MeSH
- Reproducibility of Results MeSH
- Chromatography, High Pressure Liquid MeSH
- Check Tag
- Humans MeSH
- Publication type
- Journal Article MeSH