Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Int J Med Sci ; 19(7): 1138-1146, 2022.
Article in English | MEDLINE | ID: mdl-35919818

ABSTRACT

Indoxyl sulfate (IS) and p-cresyl sulfate (PCS), protein-bound uremic toxins, can induce oxidative stress and cause renal disease progression. However, the different cytotoxic effects on renal cells between IS and PCS are not stated. Due to uremic toxins are generally found in CKD patients, the mechanisms of uremic toxins-induced renal injury are required to study. Curcumin has anti-oxidant, anti-inflammatory and anti-apoptotic effects which may be potential used to protect against renal damage. In contrast, curcumin also exert cytotoxic effects on various cells. In addition, curcumin may reduce or enhance cytotoxicity combined with different chemicals treatments. However, whether curcumin may influence uremic toxins-induced renal injury is unclear. The goal of this study is to compare the different cytotoxic effects on renal cells between IS and PCS treatment, as well as the synergistic or antagonistic effects by combination treatments with curcumin and PCS. Our experimental result shows the PCS exerts a stronger antiproliferative effect on renal tubular cells than IS treatment. In addition, our study firstly demonstrates that curcumin enhances PCS-induced cell cytotoxicity through caspase-dependent apoptotic pathway and cell cycle alteration.


Subject(s)
Curcumin , Renal Insufficiency, Chronic , Cresols/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Humans , Indican/metabolism , Indican/toxicity , Kidney/metabolism , Renal Insufficiency, Chronic/metabolism , Sulfates , Sulfuric Acid Esters/metabolism , Sulfuric Acid Esters/toxicity
2.
Toxicol Lett ; 367: 19-31, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35839976

ABSTRACT

Uremic cardiomyopathy (UCM) is a common complication in patients with chronic kidney disease (CKD) and an important risk factor for death. P-Cresyl sulfate (PCS) is a damaging factor in UCM, and Klotho is a protective factor. However, the molecular mechanisms of Klotho and PCS in UCM and the relationship between PCS and Klotho are unclear. In vitro, Klotho treatment inhibited PCS-induced cardiomyocyte hypertrophy and apoptosis by blocking mTOR phosphorylation and inhibiting DNA double-strand breaks (DSBs), respectively. Moreover, PCS increased SIRT6 protein ubiquitination and downregulated SIRT6 protein expression, while Klotho inhibited SIRT6 protein ubiquitination and upregulated SIRT6 protein expression. In a mouse model of 5/6 nephrectomy (5/6Nx)-induced UCM, the expression of Klotho in the kidney and serum was decreased, and the expression of SIRT6 protein in myocardial tissues was lower. PCS further reduced Klotho and SIRT6 expression, aggravated heart structure and function abnormalities, and increased myocardial cell apoptosis in UCM mice. Administration of Klotho protein inhibited the downregulation of SIRT6 protein expression and improved cardiac structure and function. Furthermore, serum PCS level was associated with the left ventricular mass (LVM) and left ventricular mass index (LVMI) in hemodialysis patients. In conclusion, the uremic toxin PCS injures cardiomyocytes via mTOR phosphorylation and DSBs, and Klotho antagonizes the damaging effects of PCS. Moreover, the SIRT6 protein plays an important role in UCM, and Klotho suppresses SIRT6 ubiquitination induced by PCS, further improves cardiac structure and function in UCM and exerts protective effects.


Subject(s)
Renal Insufficiency, Chronic , Sirtuins , Animals , Cresols/toxicity , Klotho Proteins , Mice , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Renal Insufficiency, Chronic/complications , Sirtuins/genetics , Sirtuins/metabolism , Sulfates/metabolism , Sulfuric Acid Esters/toxicity , TOR Serine-Threonine Kinases/metabolism , Ubiquitination
3.
J Oleo Sci ; 70(8): 1027-1037, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34248098

ABSTRACT

Liquid detergent has an increasing demand in North America, Western Europe, and Southeast Asia countries owing to its convenience to use and efficiency to clean. Alpha methyl ester sulfonates (α-MES), an anionic surfactant derived from palm oil based methyl ester, was reported to have lower manufacturing cost, good detergency with less dosage, excellent biodegradability, higher tolerance to hard water, and lower eco-toxicity as compared to linear alkylbenzene sulfonates (LABS). LABS was known as the workhorse of the detergent industry in the 20th century. Although palm-based α-MES was successfully used as the sole surfactant in powder detergent, there are still some unsettled technical issues related to phase stability and viscosity when using this anionic surfactant in heavy-duty laundry liquid detergent formulations. This paper will review not only the market overview of detergents, the application and performance of green surfactants in laundry detergents but also will highlight the technical issues related to the application of palm-based α-MES in laundry liquid detergent and some of the possible methods to overcome the formulation adversities.


Subject(s)
Alkanesulfonates/chemistry , Detergents/chemistry , Surface-Active Agents/chemistry , Alkanesulfonates/toxicity , Animals , Biodegradation, Environmental , Detergents/toxicity , Esters/chemistry , Esters/toxicity , Glycolipids/chemistry , Glycolipids/toxicity , Green Chemistry Technology , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/toxicity , Surface-Active Agents/toxicity , Viscosity
4.
Methods Mol Biol ; 2325: 215-227, 2021.
Article in English | MEDLINE | ID: mdl-34053061

ABSTRACT

Protein-bound uremic toxins (PBUTs) are bioactive microbiota metabolites originated exclusively from protein fermentation of the bacterial community resident within the gut microbiota, whose composition and function is profoundly different in the chronic kidney disease (CKD) population. PBUTs accumulate in the later stages of CKD because they cannot be efficiently removed by conventional hemodialysis due to their high binding affinity for albumin, worsening their toxic effects, especially at the cardiovascular level. The accumulation of uremic toxins, along with oxidative stress products and pro-inflammatory cytokines, characterizes the uremic status of CKD patients which is increasingly associated to a state of immune dysfunction including both immune activation and immunodepression. Furthermore, the links between immune activation and cardiovascular disease (CVD), and between immunodepression and infection diseases, which are the two major complications of CKD, are becoming more and more evident. This review summarizes and discusses the current state of knowledge on the role of the main PBUTs, namely indoxyl sulfate and p-cresyl sulfate, as regulators of immune response in CKD, in order to understand whether a microbiota modulation may be useful in the management of its main complications, CVD, and infections. Summarizing the direct effects of PBUT on immune system we may conclude that PCS seemed to be associated to an immune deficiency status of CKD mainly related to the adaptative immune response, while IS seemed to reflect the activation of both innate and adaptative immune systems likely responsible of the CKD-associated inflammation. However, the exact role of IS and PCS on immunity modulation in physiological and pathological state still needs in-depth investigation, particularly in vivo studies.


Subject(s)
Cresols/toxicity , Indican/toxicity , Renal Insufficiency, Chronic/immunology , Sulfuric Acid Esters/toxicity , T-Lymphocytes/immunology , Toxins, Biological/urine , Uremia/immunology , Adaptive Immunity , Cardiovascular Diseases/complications , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/urine , Cresols/metabolism , Gastrointestinal Microbiome/immunology , Humans , Immunity, Innate , Indican/metabolism , Inflammation/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/urine , Sulfuric Acid Esters/metabolism , Uremia/metabolism , Uremia/urine
5.
Toxicol Lett ; 347: 12-22, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33945863

ABSTRACT

p-Cresyl sulfate (PCS), indoxyl sulfate (IS), and inorganic phosphate (Pi) are uremic toxins found in chronic kidney disease (CKD) that are closely related to endothelial extracellular vesicles (EVs) formation. The present study aimed to understand the role of EVs and their role in cell adhesion and migration, inflammation, and oxidative stress. Human endothelial cells were treated with PCS, IS, and Pi in pre-established uremic and kinetic recommendations. EVs were characterized using scanning electron microscopy, flow cytometry, and NanoSight assays. The concentrations of EVs were established using Alamar Blue and MTT assays. Cell adhesion to extracellular matrix proteins was analyzed using an adhesion assay. Inflammation and oxidative stress were assessed by vascular cell adhesion molecule-1 expression/monocyte migration and reactive oxygen species production, respectively. The capacity of EVs to stimulate endothelial cell migration was evaluated using a wound-healing assay. Our data showed that endothelial cells stimulated with uremic toxins can induce the formation of EVs of different sizes, quantities, and concentrations, depending on the uremic toxin used. Cell adhesion was significantly (P < 0.01) stimulated in cells exposed to PCS-induced extracellular vesicles (PCSEVs) and inorganic phosphate-induced extracellular vesicles (PiEVs). Cell migration was significantly (P < 0.05) stimulated by PCSEVs. VCAM-1 expression was evident in cells treated with PCSEVs and IS-induced extracellular vesicles (ISEVs). EVs are not able to stimulate monocyte migration or oxidative stress. In conclusion, EVs may be a biomarker of endothelial injury and the inflammatory process, playing an important role in cell-to-cell communication and pathophysiological processes, although more studies are needed to better understand the mechanisms of EVs in uremia.


Subject(s)
Cell Adhesion/drug effects , Cell Movement/drug effects , Cresols/toxicity , Endothelial Cells/drug effects , Extracellular Vesicles/drug effects , Indican/toxicity , Inflammation Mediators/metabolism , Oxidative Stress/drug effects , Phosphates/toxicity , Sulfuric Acid Esters/toxicity , Uremia/pathology , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Humans , Signal Transduction , Uremia/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
6.
Toxicol Appl Pharmacol ; 425: 115553, 2021 08 15.
Article in English | MEDLINE | ID: mdl-33915121

ABSTRACT

p-Cresol sulfate, the primary metabolite of p-cresol, is a uremic toxin that has been associated with toxicities and mortalities. The study objectives were to i) characterize the contributions of human sulfotransferases (SULT) catalyzing p-cresol sulfate formation using multiple recombinant SULT enzymes (including the polymorphic variant SULT1A1*2), pooled human liver cytosols, and pooled human kidney cytosols; and ii) determine the potencies and mechanisms of therapeutic inhibitors capable of attenuating the production of p-cresol sulfate. Human recombinant SULT1A1 was the primary enzyme responsible for the formation of p-cresol sulfate (Km = 0.19 ±â€¯0.02 µM [with atypical kinetic behavior at lower substrate concentrations; see text discussion], Vmax = 789.5 ±â€¯101.7 nmol/mg/min, Ksi = 2458.0 ±â€¯332.8 µM, mean ±â€¯standard deviation, n = 3), while SULT1A3, SULT1B1, SULT1E1, and SULT2A1 contributed negligible or minor roles at toxic p-cresol concentrations. Moreover, human recombinant SULT1A1*2 exhibited reduced enzyme activities (Km = 81.5 ±â€¯31.4 µM, Vmax = 230.6 ±â€¯17.7 nmol/mg/min, Ksi = 986.0 ±â€¯434.4 µM) compared to the wild type. The sulfonation of p-cresol was characterized by Michaelis-Menten kinetics in liver cytosols (Km = 14.8 ±â€¯3.4 µM, Vmax = 1.5 ±â€¯0.2 nmol/mg/min) and substrate inhibition in kidney cytosols (Km = 0.29 ±â€¯0.02 µM, Vmax = 0.19 ±â€¯0.05 nmol/mg/min, Ksi = 911.7 ±â€¯278.4 µM). Of the 14 investigated therapeutic inhibitors, mefenamic acid (Ki = 2.4 ±â€¯0.1 nM [liver], Ki = 1.2 ±â€¯0.3 nM [kidney]) was the most potent in reducing the formation of p-cresol sulfate, exhibiting noncompetitive inhibition in human liver cytosols and recombinant SULT1A1, and mixed inhibition in human kidney cytosols. Our novel findings indicated that SULT1A1 contributed an important role in p-cresol sulfonation (hence it can be considered a probe reaction) in liver and kidneys, and mefenamic acid may be utilized as a potential therapeutic agent to attenuate the generation of p-cresol sulfate as an approach to detoxification.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cresols/metabolism , Cresols/toxicity , Mefenamic Acid/pharmacology , Sulfotransferases/metabolism , Sulfuric Acid Esters/metabolism , Sulfuric Acid Esters/toxicity , Catalysis , Cytosol/enzymology , Humans , Kidney , Liver , Recombinant Proteins , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/genetics
7.
Angew Chem Int Ed Engl ; 59(26): 10292-10296, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32243047

ABSTRACT

Elimination of pulmonary Pseudomonas aeruginosa (PA) infections is challenging to accomplish with antibiotic therapies, mainly due to resistance mechanisms. Quorum sensing inhibitors (QSIs) interfering with biofilm formation can thus complement antibiotics. For simultaneous and improved delivery of both active agents to the infection sites, self-assembling nanoparticles of a newly synthesized squalenyl hydrogen sulfate (SqNPs) were prepared. These nanocarriers allowed for remarkably high loading capacities of hydrophilic antibiotic tobramycin (Tob) and a novel lipophilic QSI at 30 % and circa 10 %, respectively. The drug-loaded SqNPs showed improved biofilm penetration and enhanced efficacy in relevant biological barriers (mucin/human tracheal mucus, biofilm), leading to complete eradication of PA biofilms at circa 16-fold lower Tob concentration than Tob alone. This study offers a viable therapy optimization and invigorates the research and development of QSIs for clinical use.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Nanoparticles/chemistry , Pseudomonas aeruginosa/drug effects , Quorum Sensing/drug effects , Tobramycin/pharmacology , Animals , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/toxicity , Drug Synergism , Humans , Microbial Sensitivity Tests , Mucus/microbiology , Nanoparticles/toxicity , Pseudomonas aeruginosa/physiology , Quinolones/pharmacology , Squalene/analogs & derivatives , Squalene/toxicity , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/toxicity , Zebrafish
8.
J Hazard Mater ; 394: 122522, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32200241

ABSTRACT

Mixed micelles of linear alkylbenzene sulfonic acid (LAS) and ether sulfate-based surfactants (SLEnS) can be added in household products and cleaning agents. SLEnS with higher ethylene oxide (EO) units in the head groups have economic and environmental advantages. This work aims to assess the influence of the number of EO units in the ecotoxicity of seven variants of SLEnS-LAS micelles (0-50 EO units) in soils. Ecotoxicological tests were carried out to assess emergence and growth of four plants species and reproduction of collembolans. Most of the variants inhibited plants growth at the highest concentrations (1237.5 µg SLEnS kg-1 of soildw). For reproduction, lower number of EO units resulted in EC50 from 924.2 (95 % CL: 760.7-1063.4) to 963.2 (95 % CL: 676.9-1249.6) µg SLEnS kg-1 of soildw, whereas for higher number of EO units (50 and 30) no inhibition was reported. Based on these results, we suggest that a higher number of EO units contribute to less hazardous formulations, confirming that different designs of surfactants may contribute to changes in the responses of terrestrial organisms. Therefore, we demonstrate that standardized ecotoxicological assays may contribute to more sustainable and effective formulations, when used upstream, prior to manufacture and marketing.


Subject(s)
Arthropods/drug effects , Micelles , Plants/drug effects , Soil Pollutants/toxicity , Surface-Active Agents/toxicity , Animals , Benzenesulfonates/chemistry , Benzenesulfonates/toxicity , Ethyl Ethers/chemistry , Ethyl Ethers/toxicity , Molecular Structure , Reproduction/drug effects , Soil Pollutants/chemistry , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/toxicity , Surface-Active Agents/chemistry
9.
Toxins (Basel) ; 12(2)2020 01 21.
Article in English | MEDLINE | ID: mdl-31973024

ABSTRACT

The goal of our study was to investigate the impact of p-cresylsulfate (PCS) on the barrier integrity in human umbilical vein endothelial cell (HUVEC) monolayers and the renal artery of chronic kidney disease (CKD) patients. We measured changes in the transendothelial electrical resistance (TEER) of HUVEC monolayers treated with PCS (0.1-0.2 mM) similar to serum levels of CKD patients. A PCS dose (0.2 mM) significantly decreased TEER over a 48-h period. Both PCS doses (0.1 and 0.2 mM) significantly decreased TEER over a 72-h period. Inter-endothelial gaps were observed in HUVECs following 48 h of PCS treatment by immunofluorescence microscopy. We also determined whether PCS induced the phosphorylation of VE-cadherin at tyrosine 658 (Y658) mediated by the phosphorylation of Src. Phosphorylated VE-cadherin (Y658) and phosphorylated Src levels were significantly higher when the cells were treated with 0.1 and 0.2 mM PCS, respectively, compared to the controls. The endothelial barrier dysfunction in the arterial intima in CKD patients was evaluated by endothelial leakage of immunoglobulin G (IgG). Increased endothelial leakage of IgG was related to the declining kidney function in CKD patients. Increased endothelial permeability induced by uremic toxins, including PCS, suggests that uremic toxins induce endothelial barrier dysfunction in CKD patients and Src-mediated phosphorylation of VE-cadherin is involved in increased endothelial permeability induced by PCS exposure.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cresols/toxicity , Endothelium, Vascular/metabolism , Renal Insufficiency, Chronic/metabolism , Sulfuric Acid Esters/toxicity , Toxins, Biological/toxicity , src-Family Kinases/metabolism , Aged , Aged, 80 and over , Cell Survival/drug effects , Glomerular Filtration Rate , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Immunoglobulin G/metabolism , Middle Aged , Permeability/drug effects , Phosphorylation , Renal Artery/metabolism , Renal Insufficiency, Chronic/physiopathology , Uremia
10.
Sci Rep ; 9(1): 3207, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824757

ABSTRACT

p-Cresyl sulfate (pCS), a uremic toxin, can cause renal damage and dysfunction. Studies suggest that renal dysfunction increases the prevalence of renal cancer. However, the effect of pCS on the proliferation and migration of renal cancer is unclear. Clear cell renal cell carcinoma (ccRCC) expresses mutant von Hippel-Lindau gene and is difficult to treat. Hypoxia-inducible factor-1α and 2-α (HIF-1α and HIF-2α) as well as microRNA-21 (miR-21) can regulate the proliferation and migration of ccRCC cells. However, the association between HIF-α and miR-21 in ccRCC remains unclear. Therefore, the effects of pCS on ccRCC cells were investigated for HIF-α and miR-21 signals. Our results showed that pCS induced overexpression of HIF-1α and promoted the proliferation and regulated epithelial-mesenchymal transition-related proteins, including E-cadherin, fibronectin, twist and vimentin in ccRCC cells. pCS treatment increased miR-21 expression. Specifically, inhibition of miR-21 blocked pCS-induced proliferation and migration. Taken together, the present results demonstrate that pCS directly induced the proliferation and migration of ccRCC cells through mechanisms involving miR-21/HIF-1α signaling pathways.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Movement/drug effects , Cresols/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/pathology , MicroRNAs/metabolism , Signal Transduction , Sulfuric Acid Esters/toxicity , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Neoplasms/genetics , MicroRNAs/genetics , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
11.
Biomed Pharmacother ; 112: 108679, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30798133

ABSTRACT

The p-cresyl sulfate accumulates in kidney disease and may be involved in renal injury. α-Lipoic acid (α-LA) acts as an antioxidant in cell injury. We investigated the effects of α-LA treatment on p-cresyl sulfate-induced renal tubular injury. p-Cresyl sulfate induced cell death, and increased Bax/Bcl-2, cleaved caspase-3, Beclin-1, and LC3BII/LC3BI in human renal proximal tubular epithelial (HK-2) cells, which was counteracted by α-LA treatment. p-Cresyl sulfate-induced apoptosis was reduced by autophagy inhibitor 3-methyladenine, and p-cresyl sulfate induced autophagy was reduced by pan-caspase inhibitor Z-VAD-FMK. Moreover, p-cresyl sulfate treatment increased the expression of ER stress proteins and decreased the expression of baculoviral IAP repeat-containing proteins 6; these effects were prevented by α-LA treatment. Apoptosis and autophagy were associated with the phosphorylation of mitogen-activated protein kinase and nuclear translocation of the nuclear factor-κB p65 subunit. Pretreatment inhibitors of p38 and JNK, and knockdown of ATF4 gene reduced apoptosis- and autophagy-related protein expressions in p-cresyl sulfate treated HK-2 cells. These results demonstrate that α-lipoic acid attenuated p-cresyl sulfate-induced cell death by suppression of apoptosis and autophagy via regulation of ER stress in HK-2 cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cresols/toxicity , Epithelial Cells/drug effects , Kidney Tubules, Proximal/drug effects , Sulfuric Acid Esters/toxicity , Thioctic Acid/pharmacology , Antioxidants/pharmacology , Apoptosis/physiology , Autophagy/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Epithelial Cells/metabolism , Humans , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/metabolism
12.
Food Chem Toxicol ; 125: 520-527, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30735752

ABSTRACT

At intestinal level, after acute or chronic exposure to iNOS-derived NO, a toxic mechanism of action leads to inflammation and degenerative diseases. The aim of this study was to investigate the effect of glucuronide and sulfate metabolites of the extra virgin olive oil phenols tyrosol (Tyr) and hydroxytyrosol (HT), in comparison with their parent compounds, on the release of NO following exposure to a pro-inflammatory stimulus, the bacterial lipopolysaccharide (LPS). Human colon adenocarcinoma cells (Caco-2), differentiated as normal enterocytes, were treated with pathological concentrations of LPS, in order to stimulate iNOS pathway, which involves NF-ĸB activation through IĸBα phosphorylation and subsequent degradation induced by Akt or MAPKs. All the tested metabolites inhibited NO release induced by LPS, acting as inhibitors of iNOS expression, with an efficacy comparable to that of the parent compounds. HT and Tyr metabolites were effective in the inhibition of IĸBα degradation. No one of the compounds was able to inhibit Akt activation, whereas they modulated p38 and ERK1/2 MAPK. Obtained data show that HT and Tyr metabolites are able to prevent a pathological NO overproduction at intestinal level, where they concentrate, thus significantly contributing to the protective activity exerted by their parent compounds against inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glucuronides/pharmacology , Nitric Oxide/antagonists & inhibitors , Phenylethyl Alcohol/analogs & derivatives , Sulfuric Acid Esters/pharmacology , Anti-Inflammatory Agents/toxicity , Caco-2 Cells , Escherichia coli/chemistry , Glucuronides/toxicity , Humans , Lipopolysaccharides/adverse effects , NF-KappaB Inhibitor alpha/metabolism , Nitric Oxide Synthase Type II/metabolism , Olea/chemistry , Olive Oil , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/toxicity , Signal Transduction/drug effects , Sulfuric Acid Esters/toxicity
13.
Nat Prod Res ; 33(20): 2977-2981, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30417660

ABSTRACT

Two new polyketides, purpurofuranone (1) and purpuropyranone (2), were isolated along with the known polyketides, cillifuranone (3) and taiwapyrone (4), from a mutant BD-3n-1 derived from the diethyl sulfate (DES) mutagenesis of a marine-derived Penicillium purpurogenum G59. The structures of 1 and 2 were elucidated by spectroscopic methods especially on the basis of X-ray diffraction and calculated optical rotations data. The plausible biosynthesis of 1 - 4 was also proposed and discussed. In preliminary MTT assay, 1 - 4 showed no notable inhibitory effects on the tested four human cancer cell lines.


Subject(s)
Mutagenesis , Penicillium/genetics , Polyketides/isolation & purification , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Mutagenesis/drug effects , Penicillium/chemistry , Polyketides/chemistry , Spectrum Analysis , Sulfuric Acid Esters/toxicity
14.
J Pharm Pharmacol ; 71(2): 251-259, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30324645

ABSTRACT

OBJECTIVES: Andrographlide sulphonate E, namely sodium 9-dehydro-17-hydro-andrographolide-19-yl sulphate, was one of the major ingredients of Xiyanping injection. The present study aimed to demonstrate its suitability as a reference standard for use of quality control of this traditional Chinese medicine preparation made from andrographlide that has been widely used to treat various infectious diseases. METHODS: The stable crystals were prepared for unambiguous elucidation of the chemical structure by comprehensive spectral and thermal analysis. The anti-inflammatory effects were investigated using in vitro and in vivo methods, and the potential allergenic risk related with safety was evaluated by in silico molecular docking analysis. KEY FINDINGS: The dihydrated sulphonate derivative could be present as orthorhombic crystals with stable three-dimensional supramolecular structure, providing it the favourable physico-chemical stability as reference substance. It exhibited potent anti-inflammatory activity both in vitro and in vivo, suggesting the potency responsible for clinic efficacy of Xiyanping. Molecular docking further demonstrated its low risk of allergic reaction, as well as the proposed mechanism of anaphylactic effect of andrographolide analogues. CONCLUSIONS: Dihydrated sodium 9-dehydro-17-hydro-andrographolide-19-yl sulphate may be the ideal reference standard for use in quality control of Xiyanping.


Subject(s)
Anaphylaxis/etiology , Anti-Inflammatory Agents/pharmacology , Diterpenes/pharmacology , Drugs, Chinese Herbal/pharmacology , Sulfuric Acid Esters/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/toxicity , Crystallization , Diterpenes/chemistry , Diterpenes/toxicity , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/toxicity , Injections , Male , Medicine, Chinese Traditional/methods , Mice , Molecular Docking Simulation , Quality Control , RAW 264.7 Cells , Reference Standards , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/toxicity
15.
Eur Rev Med Pharmacol Sci ; 22(21): 7533-7542, 2018 11.
Article in English | MEDLINE | ID: mdl-30468503

ABSTRACT

OBJECTIVE: The study aimed to explore the effects of p-cresyl sulfate (PCS) of damaging vascular endothelial cells and promoting the formation of atherosclerosis in mice. MATERIALS AND METHODS: The apolipoprotein E (ApoE)-/- mice were fed normally and with a high-fat diet; the ApoE-/- mice fed with high-fat diet were divided into two groups and treated with blank control and PCS, respectively. The aortic arch in each group was taken and underwent the oil red O staining, and the serum PCS content in each group was detected. The basic components of plaque were observed, including foam cells, lipid deposition, and cholesterol crystal. Moreover, human umbilical vein endothelial cells were cultured and divided into control group, PCS treatment group (PCS), PCS treatment with TLR4 overexpression group (PCS+TLR4+), and PCS treatment with TLR4 knock-out group (PCS+TLR4-). The degree of endothelial cell damage was detected using a cluster of differentiation CD42b-/CD31+ endothelial microparticles (EMPs), and expressions of Toll-like receptor 4 (TLR4), triggering receptor expressed on myeloid cells-1 (TREM-1), phosphorylated-endothelial nitric oxide synthase (p-eNOS), and tumor necrosis factor-α (TNF-α) in cells were detected via Polymerase Chain Reaction (PCR) and Western blotting. RESULTS: The serum PCS concentration in high-fat ApoE-/- mice was increased, and the aortic arch sections of ApoE-/- mice treated with PCS displayed the evident atherosclerotic plaques. Experimental results of human umbilical vein endothelial cells showed that the activity of human umbilical vein endothelial cells treated with PCS declined, the expression levels of TLR4, TREM-1, and TNF-α were increased, while that of p-eNOS was decreased. After the TLR4 knockout, the above effects of PCS were reversed. CONCLUSIONS: PCS damages vascular endothelial cells through TRL4/TREM-1, thereby accelerating the formation of atherosclerosis.


Subject(s)
Atherosclerosis/chemically induced , Cresols/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Sulfuric Acid Esters/toxicity , Toll-Like Receptor 4/physiology , Triggering Receptor Expressed on Myeloid Cells-1/physiology , Animals , Cells, Cultured , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Nitric Oxide Synthase Type III/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Eur Rev Med Pharmacol Sci ; 22(18): 6149-6155, 2018 09.
Article in English | MEDLINE | ID: mdl-30280803

ABSTRACT

OBJECTIVE: To study the protective effect of simvastatin on arterial plaque instability induced by p-cresyl sulfate (PCS). MATERIALS AND METHODS: Apolipoprotein E (ApoE)-/- mice were selected as objects of this study. All mice were randomly divided into three groups: 1) the control group, 2) the PCS group and 3) the PCS + simvastatin group. After successful modeling, the levels of plasma cholesterol, triglyceride, low-density lipoprotein, high-density lipoprotein, interleukin-1 (IL-1), interleukin-6 (IL-6) and tumor necrosis factor-ß (TNF-ß) were detected. The gross specimen of coronary artery was stained. Meanwhile, oil red O staining and Sirius red staining were performed for coronary arterial sections to observe the lipid and collagen components. The expression levels of smooth muscle cells and macrophages were observed by immunohistochemistry. In addition, the expression levels of matrix metalloproteinases (MMPs), tissue inhibitors of metalloproteinases (TIMPs) and monocyte chemoattractant protein-1 (MCP-1) in tissues were detected by Western blotting. RESULTS: Simvastatin could improve atherosclerotic plaque growth and atherosclerotic plaque instability induced by PCS. Moreover, simvastatin could also improve the changes of MMPs and TIMPs caused by PCS as well as the inflammatory status in mice. CONCLUSIONS: Simvastatin can improve the inflammatory status in mice, eventually improving the arterial plaque instability caused by PCS.


Subject(s)
Cresols/toxicity , Plaque, Atherosclerotic/drug therapy , Simvastatin/therapeutic use , Sulfuric Acid Esters/toxicity , Animals , Lipids/blood , Male , Matrix Metalloproteinases/metabolism , Mice , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Protective Agents/therapeutic use , Simvastatin/pharmacology
17.
Toxicol Appl Pharmacol ; 342: 50-59, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29407365

ABSTRACT

Infection is a major cause of mortality in chronic kidney disease (CKD) patients. Although immune dysfunction is a risk factor for infection in CKD patients, its causes are not fully elucidated. In the present study, we evaluated whether p-cresyl sulfate (pCS), an intestinal bacteria-derived uremic toxin, was involved in immune dysfunction in CKD. We used osmotic pumps to establish adenine-induced renal dysfunction mice with a chronically high blood pCS concentration. Analysis of lymphocyte subsets revealed that pCS significantly reduced peripheral B cells in renal dysfunction mice. In vitro, pCS inhibited interleukin (IL)-7-induced proliferation of CD43+ B-cell progenitors and suppressed IL-7-induced phosphorylation of signal transducer and activator of transcription 5 (STAT5) in these cells. Cell cycle analysis showed that pCS significantly decreased the percentage of CD43+ B-cell progenitors in S phase and increased that in G1 phase. These results suggest that pCS suppressed IL-7-induced STAT5 signaling and inhibited B-cell progenitor proliferation, leading to reduction of peripheral B cells in adenine-induced renal dysfunction mice. Therefore, pCS decreases peripheral B cells by inhibiting proliferation of CD43+ B-cell progenitors and is a likely cause of immune dysfunction in CKD patients.


Subject(s)
Adenine/toxicity , B-Lymphocytes/pathology , Cresols/toxicity , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Sulfuric Acid Esters/toxicity , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Female , Mice , Mice, Inbred BALB C , Renal Insufficiency, Chronic/immunology
18.
PLoS One ; 13(2): e0193342, 2018.
Article in English | MEDLINE | ID: mdl-29474405

ABSTRACT

In chronic kidney disease patients, oxidative stress is generally associated with disease progression and pathogenesis of its comorbidities. Phenyl sulfate is a protein-bound uremic solute, which accumulates in chronic kidney disease patients, but little is known about its nature. Although many reports revealed that protein-bound uremic solutes induce reactive oxygen species production, the effects of these solutes on anti-oxidant level have not been well studied. Therefore, we examined the effects of protein-bound uremic solutes on glutathione levels. As a result, indoxyl sulfate, phenyl sulfate, and p-cresyl sulfate decreased glutathione levels in porcine renal tubular cells. Next we examined whether phenyl sulfate-treated cells becomes vulnerable to oxidative stress. In phenyl sulfate-treated cells, hydrogen peroxide induced higher rates of cell death than in control cells. Buthionine sulfoximine, which is known to decrease glutathione level, well mimicked the effect of phenyl sulfate. Finally, we evaluated a mixture of indoxyl sulfate, phenyl sulfate, and p-cresyl sulfate at concentrations comparable to the serum concentrations of hemodialysis patients, and we confirmed its decreasing effect on glutathione level. In conclusion, indoxyl sulfate, phenyl sulfate, and p-cresyl sulfate decrease glutathione levels, rendering the cells vulnerable to oxidative stress.


Subject(s)
Cresols/metabolism , Glutathione/metabolism , Indican/metabolism , Kidney Tubules/metabolism , Oxidative Stress/physiology , Sulfuric Acid Esters/metabolism , Animals , Antimetabolites/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Buthionine Sulfoximine/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cresols/toxicity , Dose-Response Relationship, Drug , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Indican/toxicity , Kidney Tubules/drug effects , Oxidative Stress/drug effects , Sulfuric Acid Esters/toxicity , Sus scrofa
19.
Artif Organs ; 42(1): 88-93, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28703401

ABSTRACT

An accumulation of protein-bound uremic toxins (PBUTs) is one of major reasons for development of uremia-related complications. We examined the PBUT removal ability of a hexadecyl-immobilized cellulose bead (HICB)-containing column for patients undergoing hemodialysis. Adsorption of indoxyl sulfate (IS), a representative PBUT, to HICBs was examined in vitro. The HICB column was used in patients undergoing hemodialysis for direct hemoperfusion with a regular hemodialyzer. The serum IS, indole acetic acid (IAA), phenyl sulfate (PhS), and p-cresyl sulfate (PCS) levels were measured before and after passing the column. HICBs adsorbed protein-free (free) IS in a dose- and time-dependent manner in vitro (55.4 ± 1.4% adsorption of 1 millimolar, 251 µg/mL, IS for 1 h). In clinical studies, passing the HICB-containing column decreased the serum level of free IS, IAA, PhS, and PCS levels significantly (by 34.4 ± 30.0%, 34.8 ± 25.4%, 28.4 ± 18.0%, and 34.9 ± 22.1%, respectively), but not protein-bound toxins in maintenance hemodialysis patients. HICBs absorbed some amount of free PBUTs, but the clinical trial to use HICB column did not show effect to reduce serum PBUTs level in hemodialysis patients. Adsorption treatment by means of direct hemoperfusion with regular hemodialysis may become an attractive blood purification treatment to increase PBUT removal when more effective materials to adsorb PBUTs selectively will be developed.


Subject(s)
Cellulose/chemistry , Hemoperfusion/methods , Kidney Failure, Chronic/therapy , Renal Dialysis/methods , Toxins, Biological/chemistry , Uremia/therapy , Adsorption , Aged , Blood Proteins/metabolism , Cresols/blood , Cresols/chemistry , Cresols/metabolism , Cresols/toxicity , Feasibility Studies , Female , Hemoperfusion/instrumentation , Humans , Indican/blood , Indican/chemistry , Indican/metabolism , Indican/toxicity , Indoleacetic Acids/blood , Indoleacetic Acids/chemistry , Indoleacetic Acids/metabolism , Indoleacetic Acids/toxicity , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , Male , Middle Aged , Porosity , Protein Binding , Renal Dialysis/instrumentation , Serum Albumin , Sulfuric Acid Esters/blood , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/metabolism , Sulfuric Acid Esters/toxicity , Toxins, Biological/blood , Toxins, Biological/metabolism , Toxins, Biological/toxicity , Uremia/blood , Uremia/etiology
20.
Environ Sci Pollut Res Int ; 24(22): 18630-18639, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28647878

ABSTRACT

Cd2+ resistance and bioaccumulation capacity were selected from parental Zygosaccharomyces rouxii (CRZ-0) while maintaining NaCl tolerance using protoplast mutagenesis technology. Ultraviolet-diethyl sulfate (UV-DES) cooperative mutagenesis, followed by preliminary screening and rescreening, was used to select the mutant strain CRZ-9. CRZ-9 grew better than CRZ-0 in YPD medium with 20 or 50 mg L-1 of Cd2+. Scanning electron microscopy observations and flow cytometry tests indicated that CRZ-9 was more effective at eliminating reactive oxygen species (ROS) generated by Cd2+, which led to less cellular structural damage and lower lethality. Furthermore, compared with CRZ-0, CRZ-9 exhibited increased potential for application with higher Cd2+ removal ratio, wider working pH range, and lower biomass dosage in Cd2+ bioaccumulation. The mutant strain CRZ-9 possessed improved Cd2+ resistance and bioaccumulation capacity and therefore is a promising strain to remove Cd2+ from wastewater.


Subject(s)
Cadmium/metabolism , Ecotoxicology/methods , Mutagenesis , Ultraviolet Rays , Zygosaccharomyces/physiology , Cadmium/toxicity , Reactive Oxygen Species/metabolism , Sodium Chloride/metabolism , Sulfuric Acid Esters/toxicity , Water Purification , Zygosaccharomyces/drug effects , Zygosaccharomyces/genetics , Zygosaccharomyces/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...