Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.365
1.
CNS Neurosci Ther ; 30(5): e14748, 2024 05.
Article En | MEDLINE | ID: mdl-38727518

AIMS: To investigate the characteristics of dynamic cerebral autoregulation (dCA) after intravenous thrombolysis (IVT) and assess the relationship between dCA and prognosis. METHODS: Patients with unilateral acute ischemic stroke receiving IVT were prospectively enrolled; those who did not were selected as controls. All patients underwent dCA measurements, by quantifying the phase difference (PD) and gain, at 1-3 and 7-10 days after stroke onset. Simultaneously, two dCA-based nomogram models were established to verify the predictive value of dCA for patients with mild-to-moderate stroke. RESULTS: Finally, 202 patients who received IVT and 238 who did not were included. IVT was positively correlated with higher PD on days 1-3 and 7-10 after stroke onset. PD values in both sides at 1-3 days after stroke onset and in the affected side at 7-10 days after onset were independent predictors of unfavorable outcomes in patients who received IVT. Additionally, in patients with mild-to-moderate stroke who received IVT, the dCA-based nomogram models significantly improved the risk predictive ability for 3-month unfavorable outcomes. CONCLUSION: IVT has a positive effect on dCA in patients with acute stroke; furthermore, dCA may be useful to predict the prognosis of patients with IVT.


Homeostasis , Ischemic Stroke , Thrombolytic Therapy , Humans , Male , Female , Aged , Middle Aged , Prognosis , Thrombolytic Therapy/methods , Homeostasis/physiology , Homeostasis/drug effects , Ischemic Stroke/drug therapy , Ischemic Stroke/physiopathology , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/therapeutic use , Cerebrovascular Circulation/physiology , Cerebrovascular Circulation/drug effects , Prospective Studies , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/therapeutic use , Administration, Intravenous , Predictive Value of Tests , Aged, 80 and over , Nomograms , Stroke/drug therapy , Stroke/physiopathology
2.
Ren Fail ; 46(1): 2350235, 2024 Dec.
Article En | MEDLINE | ID: mdl-38721924

Increasing evidence suggests that peritoneal fibrosis induced by peritoneal dialysis (PD) is linked to oxidative stress. However, there are currently no effective interventions for peritoneal fibrosis. In the present study, we explored whether adding caffeic acid phenethyl ester (CAPE) to peritoneal dialysis fluid (PDF) improved peritoneal fibrosis caused by PD and explored the molecular mechanism. We established a peritoneal fibrosis model in Sprague-Dawley rats through intraperitoneal injection of PDF and lipopolysaccharide (LPS). Rats in the PD group showed increased peritoneal thickness, submesothelial collagen deposition, and the expression of TGFß1 and α-SMA. Adding CAPE to PDF significantly inhibited PD-induced submesothelial thickening, reduced TGFß1 and α-SMA expression, alleviated peritoneal fibrosis, and improved the peritoneal ultrafiltration function. In vitro, peritoneal mesothelial cells (PMCs) treated with PDF showed inhibition of the AMPK/SIRT1 pathway, mitochondrial membrane potential depolarization, overproduction of mitochondrial reactive oxygen species (ROS), decreased ATP synthesis, and induction of mesothelial-mesenchymal transition (MMT). CAPE activated the AMPK/SIRT1 pathway, thereby inhibiting mitochondrial membrane potential depolarization, reducing mitochondrial ROS generation, and maintaining ATP synthesis. However, the beneficial effects of CAPE were counteracted by an AMPK inhibitor and siSIRT1. Our results suggest that CAPE maintains mitochondrial homeostasis by upregulating the AMPK/SIRT1 pathway, which alleviates oxidative stress and MMT, thereby mitigating the damage to the peritoneal structure and function caused by PD. These findings suggest that adding CAPE to PDF may prevent and treat peritoneal fibrosis.


AMP-Activated Protein Kinases , Caffeic Acids , Peritoneal Dialysis , Peritoneal Fibrosis , Phenylethyl Alcohol , Rats, Sprague-Dawley , Sirtuin 1 , Animals , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/prevention & control , Sirtuin 1/metabolism , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , Rats , Male , AMP-Activated Protein Kinases/metabolism , Peritoneal Dialysis/adverse effects , Mitochondria/drug effects , Mitochondria/metabolism , Disease Models, Animal , Signal Transduction/drug effects , Peritoneum/pathology , Peritoneum/drug effects , Peritoneum/metabolism , Homeostasis/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , Membrane Potential, Mitochondrial/drug effects , Dialysis Solutions
3.
Signal Transduct Target Ther ; 9(1): 133, 2024 May 15.
Article En | MEDLINE | ID: mdl-38744811

Sirtuin 3 (SIRT3) is well known as a conserved nicotinamide adenine dinucleotide+ (NAD+)-dependent deacetylase located in the mitochondria that may regulate oxidative stress, catabolism and ATP production. Accumulating evidence has recently revealed that SIRT3 plays its critical roles in cardiac fibrosis, myocardial fibrosis and even heart failure (HF), through its deacetylation modifications. Accordingly, discovery of SIRT3 activators and elucidating their underlying mechanisms of HF should be urgently needed. Herein, we identified a new small-molecule activator of SIRT3 (named 2-APQC) by the structure-based drug designing strategy. 2-APQC was shown to alleviate isoproterenol (ISO)-induced cardiac hypertrophy and myocardial fibrosis in vitro and in vivo rat models. Importantly, in SIRT3 knockout mice, 2-APQC could not relieve HF, suggesting that 2-APQC is dependent on SIRT3 for its protective role. Mechanically, 2-APQC was found to inhibit the mammalian target of rapamycin (mTOR)-p70 ribosomal protein S6 kinase (p70S6K), c-jun N-terminal kinase (JNK) and transforming growth factor-ß (TGF-ß)/ small mother against decapentaplegic 3 (Smad3) pathways to improve ISO-induced cardiac hypertrophy and myocardial fibrosis. Based upon RNA-seq analyses, we demonstrated that SIRT3-pyrroline-5-carboxylate reductase 1 (PYCR1) axis was closely assoiated with HF. By activating PYCR1, 2-APQC was shown to enhance mitochondrial proline metabolism, inhibited reactive oxygen species (ROS)-p38 mitogen activated protein kinase (p38MAPK) pathway and thereby protecting against ISO-induced mitochondrialoxidative damage. Moreover, activation of SIRT3 by 2-APQC could facilitate AMP-activated protein kinase (AMPK)-Parkin axis to inhibit ISO-induced necrosis. Together, our results demonstrate that 2-APQC is a targeted SIRT3 activator that alleviates myocardial hypertrophy and fibrosis by regulating mitochondrial homeostasis, which may provide a new clue on exploiting a promising drug candidate for the future HF therapeutics.


Cardiomegaly , Fibrosis , Sirtuin 3 , Animals , Sirtuin 3/genetics , Sirtuin 3/metabolism , Cardiomegaly/genetics , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Fibrosis/genetics , Rats , Mice , Isoproterenol , Humans , Mice, Knockout , Homeostasis/drug effects , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/pathology , Mitochondria/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardium/pathology , Myocardium/metabolism , Male
4.
Eur J Med Res ; 29(1): 271, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711117

Dexmedetomidine (Dex) has been used in surgery to improve patients' postoperative cognitive function. However, the role of Dex in stress-induced anxiety-like behaviors and cognitive impairment is still unclear. In this study, we tested the role of Dex in anxiety-like behavior and cognitive impairment induced by acute restrictive stress and analyzed the alterations of the intestinal flora to explore the possible mechanism. Behavioral and cognitive tests, including open field test, elevated plus-maze test, novel object recognition test, and Barnes maze test, were performed. Intestinal gut Microbe 16S rRNA sequencing was analyzed. We found that intraperitoneal injection of Dex significantly improved acute restrictive stress-induced anxiety-like behavior, recognition, and memory impairment. After habituation in the environment, mice (male, 8 weeks, 18-23 g) were randomly divided into a control group (control, N = 10), dexmedetomidine group (Dex, N = 10), AS with normal saline group (AS + NS, N = 10) and AS with dexmedetomidine group (AS + Dex, N = 10). By the analysis of intestinal flora, we found that acute stress caused intestinal flora disorder in mice. Dex intervention changed the composition of the intestinal flora of acute stress mice, stabilized the ecology of the intestinal flora, and significantly increased the levels of Blautia (A genus of anaerobic bacteria) and Coprobacillus. These findings suggest that Dex attenuates acute stress-impaired learning and memory in mice by maintaining the homeostasis of intestinal flora.


Dexmedetomidine , Gastrointestinal Microbiome , Homeostasis , Stress, Psychological , Animals , Dexmedetomidine/pharmacology , Gastrointestinal Microbiome/drug effects , Mice , Male , Homeostasis/drug effects , Stress, Psychological/complications , Stress, Psychological/drug therapy , Memory/drug effects , Memory Disorders/drug therapy , Maze Learning/drug effects , Anxiety/drug therapy
5.
Nutrients ; 16(9)2024 Apr 28.
Article En | MEDLINE | ID: mdl-38732567

Imbalances in lipid uptake and efflux and inflammation are major contributors to foam cell formation, which is considered a therapeutic target to protect against atherosclerosis. Naringin, a citrus flavonoid abundant in citrus fruits, has been reported to exert an antiatherogenic function, but its pharmacological mechanism is unclear. Naringin treatment effectively inhibits foam cell formation in THP-1 and RAW264.7 macrophages. In this study, mechanically, naringin maintained lipid homeostasis within macrophages through downregulation of the key genes for lipid uptake (MSR1 and CD36) and the upregulation of ABCA1, ABCG1 and SR-B1, which are responsible for cholesterol efflux. Meanwhile, naringin significantly decreased the cholesterol synthesis-related genes and increased the genes involved in cholesterol metabolism. Subsequently, the results showed that ox-LDL-induced macrophage inflammatory responses were inhibited by naringin by reducing the proinflammatory cytokines IL-1ß, IL-6 and TNF-α, and increasing the anti- inflammatory cytokine IL-10, which was further verified by the downregulation of pro-inflammatory and chemokine-related genes. Additionally, we found that naringin reprogrammed the metabolic phenotypes of macrophages by suppressing glycolysis and promoting lipid oxidation metabolism to restore macrophage phenotypes and functions. These results suggest that naringin is a potential drug for the treatment of AS as it inhibits macrophage foam cell formation by regulating metabolic phenotypes and inflammation.


Flavanones , Foam Cells , Homeostasis , Lipid Metabolism , Phenotype , Foam Cells/drug effects , Foam Cells/metabolism , Flavanones/pharmacology , Mice , Lipid Metabolism/drug effects , Animals , Humans , Homeostasis/drug effects , RAW 264.7 Cells , Cytokines/metabolism , Cholesterol/metabolism , THP-1 Cells , Macrophages/drug effects , Macrophages/metabolism , Lipoproteins, LDL/metabolism , Inflammation/metabolism , Inflammation/drug therapy
6.
Eur J Histochem ; 68(2)2024 May 13.
Article En | MEDLINE | ID: mdl-38742403

Chronic kidney disease (CKD) is a leading public health issue associated with high morbidity worldwide. However, there are only a few effective therapeutic strategies for CKD. Emodin, an anthraquinone compound from rhubarb, can inhibit fibrosis in tissues and cells. Our study aims to investigate the antifibrotic effect of emodin and the underlying molecular mechanism. A unilateral ureteral obstruction (UUO)-induced rat model was established to evaluate the effect of emodin on renal fibrosis development. Hematoxylin and eosin staining, Masson's trichrome staining, and immunohistochemistry staining were performed to analyze histopathological changes and fibrotic features after emodin treatment. Subsequently, a transforming growth factor-beta 1 (TGF-ß1)-induced cell model was used to assess the inhibition of emodin on cell fibrosis in vitro. Furthermore, Western blot analysis and real-time quantitative reverse transcription-polymerase chain reaction were performed to validate the regulatory mechanism of emodin on renal fibrosis progression. As a result, emodin significantly improved histopathological abnormalities in rats with UUO. The expression of fibrosis biomarkers and mitochondrial biogenesis-related proteins also decreased after emodin treatment. Moreover, emodin blocked TGF-ß1-induced fibrotic phenotype, lipid accumulation, and mitochondrial homeostasis in NRK-52E cells. Conversely, peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) silencing significantly reversed these features in emodin-treated cells. Collectively, emodin plays an important role in regulating PGC-1α-mediated mitochondria function and energy homeostasis. This indicates that emodin exhibits great inhibition against renal fibrosis and acts as a promising inhibitor of CKD.


Emodin , Fibrosis , Mitochondria , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Renal Insufficiency, Chronic , Animals , Emodin/pharmacology , Emodin/therapeutic use , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Fibrosis/drug therapy , Mitochondria/drug effects , Mitochondria/metabolism , Male , Rats , Rats, Sprague-Dawley , Homeostasis/drug effects , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Ureteral Obstruction/pathology , Ureteral Obstruction/drug therapy , Transforming Growth Factor beta1/metabolism , Cell Line
7.
Molecules ; 29(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38731579

Trehalose is a naturally occurring, non-reducing saccharide widely distributed in nature. Over the years, research on trehalose has revealed that this initially thought simple storage molecule is a multifunctional and multitasking compound protecting cells against various stress factors. This review presents data on the role of trehalose in maintaining cellular homeostasis under stress conditions and in the virulence of bacteria and fungi. Numerous studies have demonstrated that trehalose acts in the cell as an osmoprotectant, chemical chaperone, free radical scavenger, carbon source, virulence factor, and metabolic regulator. The increasingly researched medical and therapeutic applications of trehalose are also discussed.


Trehalose , Trehalose/pharmacology , Trehalose/metabolism , Humans , Animals , Fungi/metabolism , Fungi/drug effects , Bacteria/metabolism , Bacteria/drug effects , Homeostasis/drug effects , Stress, Physiological/drug effects
8.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732122

Osteoarthritis is more prevalent than any other form of arthritis and is characterized by the progressive mechanical deterioration of joints. Glucosamine, an amino monosaccharide, has been used for over fifty years as a dietary supplement to alleviate osteoarthritis-related discomfort. Silibinin, extracted from milk thistle, modifies the degree of glycosylation of target proteins, making it an essential component in the treatment of various diseases. In this study, we aimed to investigate the functional roles of glucosamine and silibinin in cartilage homeostasis using the TC28a2 cell line. Western blots showed that glucosamine suppressed the N-glycosylation of the gp130, EGFR, and N-cadherin proteins. Furthermore, both glucosamine and silibinin differentially decreased and increased target proteins such as gp130, Snail, and KLF4 in TC28a2 cells. We observed that both compounds dose-dependently induced the proliferation of TC28a2 cells. Our MitoSOX and DCFH-DA dye data showed that 1 µM glucosamine suppressed mitochondrial reactive oxygen species (ROS) generation and induced cytosol ROS generation, whereas silibinin induced both mitochondrial and cytosol ROS generation in TC28a2 cells. Our JC-1 data showed that glucosamine increased red aggregates, resulting in an increase in the red/green fluorescence intensity ratio, while all the tested silibinin concentrations increased the green monomers, resulting in decreases in the red/green ratio. We observed increasing subG1 and S populations and decreasing G1 and G2/M populations with increasing amounts of glucosamine, while increasing amounts of silibinin led to increases in subG1, S, and G2/M populations and decreases in G1 populations in TC28a2 cells. MTT data showed that both glucosamine and silibinin induced cytotoxicity in TC28a2 cells in a dose-dependent manner. Regarding endoplasmic reticulum stress, both compounds induced the expression of CHOP and increased the level of p-eIF2α/eIF2α. With respect to O-GlcNAcylation status, glucosamine and silibinin both reduced the levels of O-GlcNAc transferase and hypoxia-inducible factor 1 alpha. Furthermore, we examined proteins and mRNAs related to these processes. In summary, our findings demonstrated that these compounds differentially modulated cellular proliferation, mitochondrial and cytosol ROS generation, the mitochondrial membrane potential, the cell cycle profile, and autophagy. Therefore, we conclude that glucosamine and silibinin not only mediate glycosylation modifications but also regulate cellular processes in human chondrocytes.


Chondrocytes , Glucosamine , Homeostasis , Kruppel-Like Factor 4 , Reactive Oxygen Species , Silybin , Glucosamine/pharmacology , Glucosamine/metabolism , Humans , Silybin/pharmacology , Glycosylation/drug effects , Chondrocytes/metabolism , Chondrocytes/drug effects , Homeostasis/drug effects , Reactive Oxygen Species/metabolism , Kruppel-Like Factor 4/metabolism , Cell Line , Cell Proliferation/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cartilage/metabolism , Cartilage/drug effects , Oxidative Stress/drug effects , Osteoarthritis/metabolism , Osteoarthritis/drug therapy
9.
J Agric Food Chem ; 72(18): 10406-10419, 2024 May 08.
Article En | MEDLINE | ID: mdl-38659208

The impact of leptin resistance on intestinal mucosal barrier integrity, appetite regulation, and hepatic lipid metabolism through the microbiota-gut-brain-liver axis has yet to be determined. Water extract of Phyllanthus emblica L. fruit (WEPE) and its bioactive compound gallic acid (GA) effectively alleviated methylglyoxal (MG)-triggered leptin resistance in vitro. Therefore, this study investigated how WEPE and GA intervention relieve leptin resistance-associated dysfunction in the intestinal mucosa, appetite, and lipid accumulation through the microbiota-gut-brain-liver axis in high-fat diet (HFD)-fed rats. The results showed that WEPE and GA significantly reduced tissues (jejunum, brain, and liver) MG-evoked leptin resistance, malondialdehyde (MDA), proinflammatory cytokines, SOCS3, orexigenic neuropeptides, and lipid accumulation through increasing leptin receptor, tight junction proteins, antimicrobial peptides, anorexigenic neuropeptides, excretion of fecal triglyceride (TG), and short-chain fatty acids (SCFAs) via a positive correlation with the Allobaculum and Bifidobacterium microbiota. These novel findings suggest that WEPE holds the potential as a functional food ingredient for alleviating obesity and its complications.


Brain , Diet, High-Fat , Fruit , Gastrointestinal Microbiome , Homeostasis , Leptin , Liver , Obesity , Phyllanthus emblica , Plant Extracts , Rats, Sprague-Dawley , Animals , Gastrointestinal Microbiome/drug effects , Rats , Male , Obesity/metabolism , Obesity/drug therapy , Obesity/microbiology , Fruit/chemistry , Liver/metabolism , Liver/drug effects , Diet, High-Fat/adverse effects , Leptin/metabolism , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Phyllanthus emblica/chemistry , Brain/metabolism , Brain/drug effects , Homeostasis/drug effects , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Appetite/drug effects , Brain-Gut Axis/drug effects , Bacteria/classification , Bacteria/metabolism , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification
10.
Eur J Pharmacol ; 973: 176585, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38636799

This study aimed to explore the effects and mechanism of action of stachydrine hydrochloride (Sta) against myocardial infarction (MI) through sarcoplasmic/endoplasmic reticulum stress-related injury. The targets of Sta against MI were screened using network pharmacology. C57BL/6 J mice after MI were treated with saline, Sta (6 or 12 mg kg-1) for 2 weeks, and adult mouse and neonatal rat cardiomyocytes (AMCMs and NRCMs) were incubated with Sta (10-4-10-6 M) under normoxia or hypoxia for 2 or 12 h, respectively. Echocardiography, Evans blue, and 2,3,5-triphenyltetrazolium chloride (TTC) staining were used for morphological and functional analyses. Endoplasmic reticulum stress (ERS), unfolded protein reaction (UPR), apoptosis signals, cardiomyocyte contraction, and Ca2+ flux were detected using transmission electron microscopy (TEM), western blotting, immunofluorescence, and sarcomere and Fluo-4 tracing. The ingredient-disease-pathway-target network revealed targets of Sta against MI were related to apoptosis, Ca2+ homeostasis and ERS. Both dosages of Sta improved heart function, decreased infarction size, and potentially increased the survival rate. Sta directly alleviated ERS and UPR and elicited less apoptosis in the border myocardium and hypoxic NRCMs. Furthermore, Sta upregulated sarcoplasmic reticulum Ca2+-ATPase 2a (SERCA2a) in both ischaemic hearts and hypoxic NRCMs, accompanied by restored sarcomere shortening, resting intracellular Ca2+, and Ca2+ reuptake time constants (Tau) in Sta-treated hypoxic ARCMs. However, 2,5-di-t-butyl-1,4-benzohydroquinone (BHQ) (25 µM), a specific SERCA inhibitor, totally abolished the beneficial effect of Sta in hypoxic cardiomyocytes. Sta protects the heart from MI by upregulating SERCA2a to maintain intracellular Ca2+ homeostasis, thus alleviating ERS-induced apoptosis.


Apoptosis , Calcium , Endoplasmic Reticulum Stress , Homeostasis , Mice, Inbred C57BL , Myocytes, Cardiac , Proline/analogs & derivatives , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Endoplasmic Reticulum Stress/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Homeostasis/drug effects , Apoptosis/drug effects , Mice , Male , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Rats , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/prevention & control , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , Unfolded Protein Response/drug effects
11.
Nat Commun ; 15(1): 3410, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38649684

Estrogen receptor α (ERα) plays a crucial role in regulating glucose and energy homeostasis during type 2 diabetes mellitus (T2DM). However, the underlying mechanisms remain incompletely understood. Here we find a ligand-independent effect of ERα on the regulation of glucose homeostasis. Deficiency of ERα in the liver impairs glucose homeostasis in male, female, and ovariectomized (OVX) female mice. Mechanistic studies reveal that ERα promotes hepatic insulin sensitivity by suppressing ubiquitination-induced IRS1 degradation. The ERα 1-280 domain mediates the ligand-independent effect of ERα on insulin sensitivity. Furthermore, we identify a peptide based on ERα 1-280 domain and find that ERα-derived peptide increases IRS1 stability and enhances insulin sensitivity. Importantly, administration of ERα-derived peptide into obese mice significantly improves glucose homeostasis and serum lipid profiles. These findings pave the way for the therapeutic intervention of T2DM by targeting the ligand-independent effect of ERα and indicate that ERα-derived peptide is a potential insulin sensitizer for the treatment of T2DM.


Diabetes Mellitus, Type 2 , Estrogen Receptor alpha , Glucose , Homeostasis , Insulin Resistance , Liver , Obesity , Animals , Female , Humans , Male , Mice , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Estrogen Receptor alpha/metabolism , Glucose/metabolism , Homeostasis/drug effects , Insulin Receptor Substrate Proteins/metabolism , Liver/metabolism , Liver/drug effects , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/drug therapy , Ovariectomy , Peptides/pharmacology , Ubiquitination/drug effects
12.
ACS Nano ; 18(18): 11778-11803, 2024 May 07.
Article En | MEDLINE | ID: mdl-38652869

Severe acute pancreatitis (AP) is a life-threatening pancreatic inflammatory disease with a high mortality rate (∼40%). Existing pharmaceutical therapies in development or in clinical trials showed insufficient treatment efficacy due to their single molecular therapeutic target, poor water solubility, short half-life, limited pancreas-targeting specificity, etc. Herein, acid-responsive hollow mesoporous Prussian blue nanoparticles wrapped with neutrophil membranes and surface modified with the N,N-dimethyl-1,3-propanediamine moiety were developed for codelivering membrane-permeable calcium chelator BAPTA-AM (BA) and trypsin activity inhibitor gabexate mesylate (Ga). In the AP mouse model, the formulation exhibited efficient recruitment at the inflammatory endothelium, trans-endothelial migration, and precise acinar cell targeting, resulting in rapid pancreatic localization and higher accumulation. A single low dose of the formulation (BA: 200 µg kg-1, Ga: 0.75 mg kg-1) significantly reduced pancreas function indicators to close to normal levels at 24 h, effectively restored the cell redox status, reduced apoptotic cell proportion, and blocked the systemic inflammatory amplified cascade, resulting in a dramatic increase in the survival rate from 58.3 to even 100%. Mechanistically, the formulation inhibited endoplasmic reticulum stress (IRE1/XBP1 and ATF4/CHOP axis) and restored impaired autophagy (Beclin-1/p62/LC3 axis), thereby preserving dying acinar cells and restoring the cellular "health status". This formulation provides an upstream therapeutic strategy with clinical translation prospects for AP management through synergistic ion homeostasis regulation and pancreatic autodigestion inhibition.


Acinar Cells , Calcium , Homeostasis , Nanomedicine , Pancreatitis , Animals , Pancreatitis/drug therapy , Pancreatitis/pathology , Pancreatitis/metabolism , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Mice , Homeostasis/drug effects , Calcium/metabolism , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Nanoparticles/chemistry , Pancreas/pathology , Pancreas/drug effects , Pancreas/metabolism , Mice, Inbred C57BL , Male , Humans
13.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article En | MEDLINE | ID: mdl-38674008

Cysteine and its derivatives, including H2S, can influence bacterial virulence and sensitivity to antibiotics. In minimal sulfate media, H2S is generated under stress to prevent excess cysteine and, together with incorporation into glutathione and export into the medium, is a mechanism of cysteine homeostasis. Here, we studied the features of cysteine homeostasis in LB medium, where the main source of sulfur is cystine, whose import can create excess cysteine inside cells. We used mutants in the mechanisms of cysteine homeostasis and a set of microbiological and biochemical methods, including the real-time monitoring of sulfide and oxygen, the determination of cysteine and glutathione (GSH), and the expression of the Fur, OxyR, and SOS regulons genes. During normal growth, the parental strain generated H2S when switching respiration to another substrate. The mutations affected the onset time, the intensity and duration of H2S production, cysteine and glutathione levels, bacterial growth and respiration rates, and the induction of defense systems. Exposure to chloramphenicol and high doses of ciprofloxacin increased cysteine content and GSH synthesis. A high inverse relationship between log CFU/mL and bacterial growth rate before ciprofloxacin addition was revealed. The study points to the important role of maintaining cysteine homeostasis during normal growth and antibiotic exposure in LB medium.


Anti-Bacterial Agents , Ciprofloxacin , Cysteine , Escherichia coli , Glutathione , Homeostasis , Cysteine/metabolism , Ciprofloxacin/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/growth & development , Homeostasis/drug effects , Glutathione/metabolism , Anti-Bacterial Agents/pharmacology , Culture Media/chemistry , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Mutation , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/drug effects
14.
Toxicology ; 504: 153802, 2024 May.
Article En | MEDLINE | ID: mdl-38604439

Etomidate (ETO) is used as an anesthetic in surgery, but it is being abused in some populations. The damage caused by long-term intake of ETO to intestinal and brain functions is not yet clear, and it remains to be determined whether the drug affects the central nervous system through the gut-brain axis. This study aimed to investigate the neurotoxic and gastrointestinal effects of ETO at doses of 1 mg/kg and 3 mg/kg in mice over 14 consecutive days. The results showed that long-term injection of ETO led to drug resistance in mice, affecting their innate preference for darkness and possibly inducing dependence on ETO. The levels of 5-hydroxytryptamine in the brain, serum, and colon decreased by 37%, 51%, and 42% respectively, while the levels of γ-aminobutyric acid reduced by 38%, 52%, and 41% respectively. H&E staining revealed that ETO reduced goblet cells in the colon and damaged the intestinal barrier. The expression of tight junction-related genes Claudin4 and ZO-1 was downregulated. The intestinal flora changed, the abundance of Akkermansia and Lactobacillus decreased by 33% and 14%, respectively, while Klebsiella increased by 18%. TUNEL results showed that high-dose ETO increased apoptotic cells in the brain. The expression of Claudin1 in the brain was downregulated. Untargeted metabolomics analysis of the colon and brain indicated that ETO caused abnormalities in glycerophospholipid metabolism. Abnormal lipid metabolism might lead to the production or accumulation of lipotoxic metabolites, causing central nervous system diseases. ETO induced changes in the intestinal flora and metabolism, further affecting the central nervous system through the gut-brain axis. The study unveiled the detrimental effects on the brain and gastrointestinal system resulting from long-term intake of ETO, which holds significant implications for comprehending the adverse impact of ETO abuse on human health.


Etomidate , Gastrointestinal Microbiome , Homeostasis , Animals , Mice , Male , Homeostasis/drug effects , Etomidate/toxicity , Gastrointestinal Microbiome/drug effects , Brain/drug effects , Brain/metabolism , Intestines/drug effects , Brain-Gut Axis/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Serotonin/metabolism
15.
Behav Brain Res ; 466: 114995, 2024 May 28.
Article En | MEDLINE | ID: mdl-38599251

Neurodegenerative disorders have a pathophysiology that heavily involves neuroinflammation. In this study, we used lipopolysaccharide (LPS) to create a model of cognitive impairment by inducing systemic and neuroinflammation in experimental animals. LPS was injected intraperitoneally at a dose of 0.5 mg/kg during the last seven days of the study. Adalimumab (ADA), a TNF-α inhibitor, was injected at a dose of 10 mg/kg a total of 3 times throughout the study. On the last two days of the experiment, 50 mg/kg of curcumin was administered orally as a positive control group. Open field (OF) and elevated plus maze tests (EPM) were used to measure anxiety-like behaviors. The tail suspension test (TST) was used to measure depression-like behaviors, while the novel object recognition test (NOR) was used to measure learning and memory activities. Blood and hippocampal TNF α and nitric oxide (NO) levels, hippocampal BDNF, CREB, and ACh levels, and AChE activity were measured by ELISA. LPS increased anxiety and depression-like behaviors while decreasing the activity of the learning-memory system. LPS exerted this effect by causing systemic and neuroinflammation, cholinergic dysfunction, and impaired BDNF release. ADA controlled LPS-induced behavioral changes and improved biochemical markers. ADA prevented cognitive impairment induced by LPS by inhibiting inflammation and regulating the release of BDNF and the cholinergic pathway.


Acetylcholine , Brain-Derived Neurotrophic Factor , Cognitive Dysfunction , Neuroinflammatory Diseases , Nitric Oxide , Sepsis , Tumor Necrosis Factor-alpha , Animals , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Mice , Brain-Derived Neurotrophic Factor/metabolism , Nitric Oxide/metabolism , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Acetylcholine/metabolism , Sepsis/complications , Sepsis/metabolism , Sepsis/drug therapy , Lipopolysaccharides/pharmacology , Adalimumab/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , Disease Models, Animal , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/etiology , Homeostasis/drug effects , Depression/metabolism , Depression/drug therapy , Depression/etiology , Behavior, Animal/drug effects , Tumor Necrosis Factor Inhibitors/pharmacology
16.
Food Funct ; 15(8): 4109-4121, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38597225

While there have been advancements in understanding the direct and indirect impact of riboflavin (B2) on intestinal inflammation, the precise mechanisms are still unknown. This study focuses on evaluating the effects of riboflavin (B2) supplementation on a colitis mouse model induced with 3% dextran sodium sulphate (DSS). We administered three different doses of oral B2 (VB2L, VB2M, and VB2H) and assessed its impact on various physiological and biochemical parameters associated with colitis. Mice given any of the three doses exhibited relative improvement in the symptoms and intestinal damage. This was evidenced by the inhibition of the pro-inflammatory cytokines TNF-α, IL-1ß, and CALP, along with an increase in the anti-inflammatory cytokine IL-10. B2 supplementation also led to a restoration of oxidative homeostasis, as indicated by a decrease in myeloperoxidase (MPO) and malondialdehyde (MDA) levels and an increase in reduced glutathione (GSH) and catalase (CAT) activities. B2 intervention showed positive effects on intestinal barrier function, confirmed by increased expression of tight junction proteins (occludin and ZO-1). B2 was linked to an elevated relative abundance of Actinobacteriota, Desulfobacterota, and Verrucomicrobiota. Notably, Verrucomicrobiota showed a significant increase in the VB2H group, reaching 15.03% relative abundance. Akkermansia exhibited a negative correlation with colitis and might be linked to anti-inflammatory function. Additionally, a remarkable increase in n-butyric acid, i-butyric acid, and i-valeric acid was reported in the VB2H group. The ameliorating role of B2 in gut inflammation can be attributed to immune system modulation as well as alterations in the gut microbiota composition, along with elevated levels of fecal SCFAs.


Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Homeostasis , Mice, Inbred C57BL , Riboflavin , Animals , Gastrointestinal Microbiome/drug effects , Mice , Colitis/drug therapy , Colitis/chemically induced , Dextran Sulfate/adverse effects , Riboflavin/pharmacology , Homeostasis/drug effects , Male , Disease Models, Animal , Cytokines/metabolism , Inflammation/drug therapy , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism
17.
Open Vet J ; 14(3): 830-839, 2024 Mar.
Article En | MEDLINE | ID: mdl-38682150

Background: Heat stress (HS) is a main abiotic stress factor for the health and welfare of animals. Recently, the use of nano-emulsion essential oils exhibited a promising approach to mitigate the detrimental impacts of abiotic and biotic stresses, ultimately contributing to the global aim of sustainable livestock production. Aim: The current study was piloted to assess the impact of eugenol nano-emulsion (EUGN) supplementation on growth performance, serum metabolites, redox homeostasis, immune response, and pro-inflammatory reactions in growing rabbits exposed to HS. Methods: A total of 100 male weaning rabbits aged 35 days were divided into 4 treatments. Rabbits were fed the diet with EUGN at different concentrations: 0 (control group; EUGN0), 50 (EUGN50), 100 (EUGN100), and 150 (EUGN150) mg/kg diet for 8 weeks under summer conditions. Results: Dietary EUGN levels significantly improved (p < 0.05) the body weight, body weight gain, carcass weights, and improved feed conversion ratio of rabbits. EUGN supplementation significantly increased Hb, platelets, and red blood cells , while the mean corpuscular hemoglobin and eosinophils were significantly decreased compared to the control one. Compared with EUGN0 stressed rabbits, all EUGN-experimental groups had a reduction in levels of total glycerides (p < 0.01), uric acid, total bilirubin, direct bilirubin, and gamma-glutamyl transpeptidase (p < 0.01). Total antioxidant capacity and glutathione peroxidase were significantly improved by EUGN treatment when compared to the control one (p < 0.01), while the EUGN100 exhibited the greatest levels of catalase. Lipid peroxidation (malondialdehyde) was significantly decreased in EUGN-treated groups. All pro-inflammatory cytokines serum interleukin 4, Interleukin 1ß, and tumor necrosis factor alpha were considerably decreased after dietary EUGN supplementation (p < 0.05). The serum concentrations of immunoglobulins (IgG and IgM) were significantly improved in rabbits of the EUGN150 group. Conclusion: This study shows that EUGN can be used as a novel feed additive to enhance the growth performance, immune variables, and antioxidants, and reduce the inflammatory response of growing rabbits exposed to thermal stress.


Animal Feed , Diet , Dietary Supplements , Eugenol , Homeostasis , Animals , Rabbits , Eugenol/administration & dosage , Eugenol/pharmacology , Male , Dietary Supplements/analysis , Animal Feed/analysis , Homeostasis/drug effects , Diet/veterinary , Oxidation-Reduction/drug effects , Emulsions , Inflammation/veterinary , Heat-Shock Response/drug effects
18.
Biomolecules ; 14(4)2024 Mar 27.
Article En | MEDLINE | ID: mdl-38672424

Originally developed as a chemotherapeutic agent, miltefosine (hexadecylphosphocholine) is an inhibitor of phosphatidylcholine synthesis with proven antiparasitic effects. It is the only oral drug approved for the treatment of Leishmaniasis and American Trypanosomiasis (Chagas disease). Although its precise mechanisms are not yet fully understood, miltefosine exhibits broad-spectrum anti-parasitic effects primarily by disrupting the intracellular Ca2+ homeostasis of the parasites while sparing the human hosts. In addition to its inhibitory effects on phosphatidylcholine synthesis and cytochrome c oxidase, miltefosine has been found to affect the unique giant mitochondria and the acidocalcisomes of parasites. Both of these crucial organelles are involved in Ca2+ regulation. Furthermore, miltefosine has the ability to activate a specific parasite Ca2+ channel that responds to sphingosine, which is different to its L-type VGCC human ortholog. Here, we aimed to provide an overview of recent advancements of the anti-parasitic mechanisms of miltefosine. We also explored its multiple molecular targets and investigated how its pleiotropic effects translate into a rational therapeutic approach for patients afflicted by Leishmaniasis and American Trypanosomiasis. Notably, miltefosine's therapeutic effect extends beyond its impact on the parasite to also positively affect the host's immune system. These findings enhance our understanding on its multi-targeted mechanism of action. Overall, this review sheds light on the intricate molecular actions of miltefosine, highlighting its potential as a promising therapeutic option against these debilitating parasitic diseases.


Calcium , Chagas Disease , Homeostasis , Leishmaniasis , Phosphorylcholine , Phosphorylcholine/analogs & derivatives , Humans , Phosphorylcholine/pharmacology , Phosphorylcholine/therapeutic use , Chagas Disease/drug therapy , Chagas Disease/parasitology , Chagas Disease/metabolism , Calcium/metabolism , Leishmaniasis/drug therapy , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Homeostasis/drug effects , Animals , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Mitochondria/metabolism , Mitochondria/drug effects , Leishmania/drug effects , Leishmania/metabolism , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/metabolism
19.
Pestic Biochem Physiol ; 201: 105904, 2024 May.
Article En | MEDLINE | ID: mdl-38685226

Arsenic (As) and polystyrene nanoplastics (PSNPs) co-exposure induced biotoxicity and ecological risks have attracted wide attention. However, the combined effects of As and PSNPs on the kidney and their underlying mechanisms of toxicities remain to be explored. Here, we investigated the effects of As and PSNPs co-exposure on structure and function in mice kidney, and further explored the possible mechanisms. In this study, we identified that co-exposure to As and PSNPs exhibited conspicuous renal structural damage and pathological changes, accompanied by renal tissue fibrosis (increased protein expression of Collagen I and α-SMA and deposition of collagen fibers), whereas alone exposure to As or PSNPs does not exhibit nephrotoxicity. Subsequently, our results further showed that combined action of As and PSNPs induced mitochondrial oxidative damage and impaired mitochondrial dynamic balance. Furthermore, co-treatment with As and PSNPs activated NCOA4-mediated ferritinophagy and ferroptosis in mice kidney and TCMK-1 cells, which was confirmed by the changes in the expression of ferritinophagy and ferroptosis related indicators (NCOA4, LC3, ATG5, ATG7, FTH1, FTL, GPX4, SLC7A11, FSP1, ACSL4 and PTGS2). Meaningfully, pretreatment with the mtROS-targeted scavenger Mito-TEMPO significantly attenuated As and PSNPs co-exposure induced mitochondrial damage, ferritinophagy and ferroptosis. In conclusion, these findings demonstrated that mtROS-dependent ferritinophagy and ferroptosis are important factors in As and PSNPs co-exposure induced kidney injury and fibrosis. This study provides a new insight into the study of combined toxicity of nanoplastics and heavy metal pollutants.


Arsenic , Ferroptosis , Kidney , Mitochondria , Polystyrenes , Animals , Ferroptosis/drug effects , Polystyrenes/toxicity , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Arsenic/toxicity , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Homeostasis/drug effects , Ferritins/metabolism , Nanoparticles/toxicity , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
20.
Int J Mol Sci ; 25(8)2024 Apr 21.
Article En | MEDLINE | ID: mdl-38674128

Type II pneumocytes are the target of the SARS-CoV-2 virus, which alters their redox homeostasis to increase reactive oxygen species (ROS). Melatonin (MT) has antioxidant proprieties and protects mitochondrial function. In this study, we evaluated whether treatment with MT compensated for the redox homeostasis alteration in serum from COVID-19 patients. We determined oxidative stress (OS) markers such as carbonyls, glutathione (GSH), total antioxidant capacity (TAC), thiols, nitrites (NO2-), lipid peroxidation (LPO), and thiol groups in serum. We also studied the enzymatic activities of glutathione peroxidase (GPx), glutathione-S-transferase (GST), reductase (GR), thioredoxin reductase (TrxR), extracellular superoxide dismutase (ecSOD) and peroxidases. There were significant increases in LPO and carbonyl quantities (p ≤ 0.03) and decreases in TAC and the quantities of NO2-, thiols, and GSH (p < 0.001) in COVID-19 patients. The activities of the antioxidant enzymes such as ecSOD, TrxR, GPx, GST, GR, and peroxidases were decreased (p ≤ 0.04) after the MT treatment. The treatment with MT favored the activity of the antioxidant enzymes that contributed to an increase in TAC and restored the lost redox homeostasis. MT also modulated glucose homeostasis, functioning as a glycolytic agent, and inhibited the Warburg effect. Thus, MT restores the redox homeostasis that is altered in COVID-19 patients and can be used as adjuvant therapy in SARS-CoV-2 infection.


Antioxidants , COVID-19 Drug Treatment , COVID-19 , Homeostasis , Melatonin , Oxidation-Reduction , Oxidative Stress , SARS-CoV-2 , Melatonin/therapeutic use , Melatonin/pharmacology , Humans , Oxidation-Reduction/drug effects , COVID-19/metabolism , COVID-19/virology , COVID-19/blood , Homeostasis/drug effects , Antioxidants/metabolism , Antioxidants/therapeutic use , Oxidative Stress/drug effects , Male , Female , Middle Aged , SARS-CoV-2/drug effects , Lipid Peroxidation/drug effects , Aged , Adult , Reactive Oxygen Species/metabolism , Glutathione/metabolism , Glutathione/blood
...