Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 442
Filter
1.
Molecules ; 26(18)2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34576927

ABSTRACT

Garden-cultivated Ginseng (GG) and mountain-cultivated Ginseng (MG) both belong to Panax Ginseng C. A. Meyer. However, the effective substances which can be used to distinguish GG from MG remain obscure. Therefore, the purpose of this study was to screen for discriminating markers that can assist in the correct identification of GG and MG. HPLC Q-TOF/MS and various chemometrics methods were used to analyze the chemical profiles of 13 batches of Ginseng and to explore the characteristic constituents of both GG and MG. The hepatocyte-protecting effects of GG and MG were investigated through a paclitaxel-induced liver injury model. Through a combination of correlation analysis and bioinformatic techniques, markers for differentiation between GG and MG were ascertained. A total of 40 and 41 compounds were identified in GG and MG, respectively, and 15 characteristic ingredients contributed significantly to the discrimination of GG from MG. Correlation analysis and network pharmacology were applied and ginsenosides Rg1, Re, Rb1, Rc, Rb2, and Rg3 were found to be discriminating markers of GG and MG. Six markers for the identification of GG and MG were screened out by a step-wise mutually oriented "chemical profiling-pharmaceutical effect" correlation strategy, which is of great significance for future quality assessment of Ginseng products.


Subject(s)
Cheminformatics/methods , Panax/chemistry , Protective Agents/chemistry , Protective Agents/pharmacology , Animals , Biomarkers, Pharmacological , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Chromatography, High Pressure Liquid , Gardens , Ginsenosides/analysis , Ginsenosides/chemistry , Mass Spectrometry , Paclitaxel/adverse effects , Panax/growth & development , Protective Agents/pharmacokinetics , Rats, Sprague-Dawley
2.
Nutrients ; 13(8)2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34444906

ABSTRACT

Diabetic kidney disease (DKD) has become a global health concern, with about 40% of people living with type 1 and type 2 diabetes mellitus developing DKD. Upregulation of vascular endothelial growth factor (VEGF) in the kidney is a significant pathology of DKD associated with increased glomerular vascular permeability. To date, however, current anti-VEGF therapies have demonstrated limited success in treating DKD. Recent studies have shown that artificial sweeteners exhibit anti-VEGF potential. The aim of this study was therefore to assess the effects of aspartame, saccharin, and sucralose on VEGF-induced leak using an in vitro model of the glomerular endothelium. Saccharin and sucralose but not aspartame protected against VEGF-induced permeability. Whilst the sweeteners had no effect on traditional VEGF signalling, GC-MS analysis demonstrated that the sweetener sucralose was not able to enter the glomerular endothelial cell to exert the protective effect. Chemical and molecular inhibition studies demonstrated that sweetener-mediated protection of the glomerular endothelium against VEGF is dependent on the sweet taste receptor, T1R3. These studies demonstrate the potential for sweeteners to exert a protective effect against VEGF-induced increased permeability to maintain a healthy endothelium and protect against vascular leak in the glomerulus in settings of DKD.


Subject(s)
Capillary Permeability/drug effects , Protective Agents/pharmacokinetics , Saccharin/pharmacokinetics , Sucrose/analogs & derivatives , Sweetening Agents/pharmacology , Aspartame/pharmacokinetics , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Endothelial Cells , Endothelium, Vascular/metabolism , Humans , In Vitro Techniques , Kidney/blood supply , Microvessels/metabolism , Sucrose/pharmacokinetics , Vascular Endothelial Growth Factors/metabolism
3.
Biomed Pharmacother ; 142: 111927, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34339914

ABSTRACT

Recent studies demonstrated that dihydromyricetin (DHM) has prominent therapeutic effects on liver injury and liver cancer. By summarizing the current preclinical in vitro and in vivo studies, the present review examines the preventive and therapeutic effects of DHM on liver disorders as well as its potential mechanisms. Briefly, in both chemical- and alcohol-induced liver injury models, DHM ameliorates hepatocyte necrosis and steatosis while promoting liver regeneration. In addition, DHM can alleviate nonalcoholic fatty liver disease (NAFLD) via regulating lipid/glucose metabolism, probably due to its anti-inflammatory or sirtuins-dependent mechanisms. Furthermore, DHM treatment inhibits cell proliferation, induces apoptosis and autophagy and regulates redox balance in liver cancer cells, thus exhibiting remarkable anti-cancer effects. The pharmacological mechanisms of DHM may be associated with its anti-inflammatory, anti-oxidative and apoptosis-regulatory benefits. With the accumulating interests in utilizing natural products to target common diseases, our work aims to improve the understanding of DHM acting as a novel drug candidate for liver diseases and to accelerate its translation from bench to bedside.


Subject(s)
Flavonols/pharmacology , Flavonols/therapeutic use , Liver Diseases/prevention & control , Protective Agents/pharmacology , Protective Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/prevention & control , Flavonols/pharmacokinetics , Humans , Liver Diseases/metabolism , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/prevention & control , Liver Failure, Acute/metabolism , Liver Failure, Acute/prevention & control , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Liver Regeneration/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Protective Agents/pharmacokinetics
4.
Mol Nutr Food Res ; 65(18): e2100253, 2021 09.
Article in English | MEDLINE | ID: mdl-34331394

ABSTRACT

SCOPE: Lactoferrin (Lf) possess a protective potential to liver, but whether it can prevent alcoholic liver injury (ALI) remains unclear. METHODS AND RESULTS: Four groups of male C57BL/6J mice are fed with different diets, namely, AIN-93G diet for control (CON) and ethanol (EtOH) groups, and AIN-93G diet with 0.4% and 4% casein replaced by Lf for low-dose Lf (LLf) and high-dose Lf (HLf) groups, respectively. ALI is induced by giving 20% ethanol ad libitum combined with four "binges". Lf can remarkably decrease EtOH-induced mortality. Lf promotes aldehyde dehydrogenase-2 (ALDH2) expression and suppressing cytochrome P450 2E1 (CYP2E1) overexpression, resulting in the reduced hepatic superoxide and inflammation levels, which ultimately leads to the hepatic injury alleviation. However, HLf increases acetyl-CoA carboxylase and fatty acid synthase protein levels, which suggests that excessive intake may weaken the beneficial effects of Lf. Moreover, LLf increases the relative abundances of Akkermansia and Lactobacillus. Additionally, the study shows that Lf likely exerts action in its digestive product forms rather than intact Lf molecular in normal condition. CONCLUSION: LLf can ameliorate ALI, which is associated with the regulation of hepatic alcohol metabolism and the modulation of gut microbiota. However, excessive Lf intake may result in a diminished benefit.


Subject(s)
Gastrointestinal Microbiome/drug effects , Lactoferrin/pharmacology , Liver Diseases, Alcoholic/prevention & control , Liver/drug effects , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Animals , Cattle , Cytochrome P-450 CYP2E1/metabolism , Gastrointestinal Microbiome/physiology , Gene Expression Regulation/drug effects , Jejunum/drug effects , Jejunum/pathology , Lactoferrin/administration & dosage , Lactoferrin/pharmacokinetics , Liver/metabolism , Liver/pathology , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/microbiology , Liver Diseases, Alcoholic/mortality , Male , Mice, Inbred C57BL , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism
5.
Pharmacology ; 106(5-6): 233-243, 2021.
Article in English | MEDLINE | ID: mdl-33849010

ABSTRACT

BACKGROUND: Our liver has a variety of vital functions including removing poisons, storing energy, immunological roles, and secretory and excretory functions. It may face some kinds of diseases caused by viruses, hepatotoxic chemicals, drugs, alcohol, and inherited disorders. Oxidative stress and inflammation are in the core of mechanisms of liver damages induced by viruses or chemical agents. SUMMARY: Morus nigra (M. nigra), generally known as black mulberry, exhibited wide-spectrum pharmacological effects including antidiabetic, antinociceptive, anticancer, and hepatoprotective activities. Different parts of this plant particularly the fruit and leaf have shown beneficial effects on hepatocytes in cell culture and animal models of liver damages induced by chemicals (e.g., CCl4), drugs (e.g., paracetamol), diet (e.g., high fat), diabetes, etc. The beneficial effects of M. nigra on the liver are attributed to the presence of considerable amounts of phenolic compounds such as anthocyanins, flavonols, and phenolic acids. The present review is aimed to focus on the hepatoprotective activities of M. nigra and its phytochemicals and the mechanisms responsible for these activities. Key Messages: The evidence reviewed in this study can help design clinical trials on M. nigra in patients with liver disorders and develop a hepatoprotective herbal medicine.


Subject(s)
Liver/drug effects , Morus/chemistry , Phenols/pharmacology , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Protective Agents/pharmacology , Animals , Humans , Liver Diseases/drug therapy , Liver Diseases/etiology , Morus/adverse effects , Phenols/adverse effects , Phenols/pharmacokinetics , Phenols/therapeutic use , Phytochemicals/adverse effects , Phytochemicals/pharmacokinetics , Phytochemicals/therapeutic use , Plant Extracts/adverse effects , Plant Extracts/pharmacokinetics , Plant Extracts/therapeutic use , Protective Agents/adverse effects , Protective Agents/pharmacokinetics , Protective Agents/therapeutic use
6.
J Pharm Pharmacol ; 73(4): 535-544, 2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33793835

ABSTRACT

OBJECTIVES: Identifying drugs with time-varying efficacy or toxicity, and understanding the underlying mechanisms would help to improve treatment efficacy and reduce adverse effects. In this study, we uncovered that the therapeutic effect of Fuzi (the lateral root of Aconitum carmichaelii Debeaux) depended on the dosing time in mice with adenine-induced chronic kidney disease (CKD). METHODS: The Fuzi efficacy was determined by biomarker measurements [i.e. plasma creatinine (CRE), blood urea nitrogen (BUN) and urinary N-acetyl-ß-D-glucosaminidase (NAG)], as well as inflammation, fibrosis and histological analyses. Circadian regulation of Fuzi pharmacokinetics and efficacy was evaluated using brain and muscle Arnt-like protein-1 (Bmal1)-deficient (Bmal1-/-) mice. KEY FINDINGS: The Fuzi efficacy was higher when the drug was dosed at ZT10 and was lower when the drug was dosed at other times (ZT2, ZT6, ZT14, ZT18 and ZT22) according to measurements of plasma CRE, BUN and urinary NAG. Consistently, ZT10 (5 PM) dosing showed a stronger protective effect on the kidney (i.e. less extensive tubular injury) as compared to ZT22 (5 AM) dosing. This was supported by lower levels of inflammatory and fibrotic factors (IL-1ß, IL-6, Tnf-α, Ccl2, Tgfb1 and Col1a1) at ZT10 than at ZT22. Pharmacokinetic analyses showed that the area under the curve (AUC) values (reflective of systemic exposure) and renal distribution of aconitine, hypaconitine and mesaconitine (three putative active constituents) for Fuzi dosing at ZT10 were significantly higher than those for herb dosing at ZT22, suggesting a role of circadian pharmacokinetics in Fuzi chronoefficacy. Drug efficacy studies confirmed that aconitine, hypaconitine and mesaconitine possessed a kidney-protecting effect. In addition, genetic knockout of Bmal1 in mice abolished the time-dependency of Fuzi pharmacokinetics and efficacy. This reinforced the existence of chronoefficacy for Fuzi and supported the role of circadian pharmacokinetics in Fuzi chronoefficacy. CONCLUSIONS: The efficacy of Fuzi against CKD depends on the dosing time in mice, which is associated with circadian pharmacokinetics of the three main active constituents (i.e. aconitine, hypaconitine and mesaconitine). These findings highlight the relevance of dosing time in the therapeutic outcomes of herbal medicines.


Subject(s)
Chronopharmacokinetics , Diterpenes , Drugs, Chinese Herbal , Renal Insufficiency, Chronic , ARNTL Transcription Factors/genetics , Aconitine/analogs & derivatives , Aconitine/analysis , Alkaloids/administration & dosage , Alkaloids/pharmacokinetics , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Diterpenes/administration & dosage , Diterpenes/pharmacokinetics , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacokinetics , Kidney Function Tests/methods , Mice , Mice, Knockout , Plant Roots , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Treatment Outcome
7.
J Endocrinol Invest ; 44(4): 819-842, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32772323

ABSTRACT

PURPOSE: In both preclinical and clinical settings, testosterone treatment (TTh) of hypogonadism has shown beneficial effects on insulin sensitivity and visceral and liver fat accumulation. This prospective, observational study was aimed at assessing the change in markers of fat and liver functioning in obese men scheduled for bariatric surgery. METHODS: Hypogonadal patients with consistent symptoms (n = 15) undergoing 27.63 ± 3.64 weeks of TTh were compared to untreated eugonadal (n = 17) or asymptomatic hypogonadal (n = 46) men. A cross-sectional analysis among the different groups was also performed, especially for data derived from liver and fat biopsies. Preadipocytes isolated from adipose tissue biopsies were used to evaluate insulin sensitivity, adipogenic potential and mitochondrial function. NAFLD was evaluated by triglyceride assay and by calculating NAFLD activity score in liver biopsies. RESULTS: In TTh-hypogonadal men, histopathological NAFLD activity and steatosis scores, as well as liver triglyceride content were lower than in untreated-hypogonadal men and comparable to eugonadal ones. TTh was also associated with a favorable hepatic expression of lipid handling-related genes. In visceral adipose tissue and preadipocytes, TTh was associated with an increased expression of lipid catabolism and mitochondrial bio-functionality markers. Preadipocytes from TTh men also exhibited a healthier morpho-functional phenotype of mitochondria and higher insulin-sensitivity compared to untreated-hypogonadal ones. CONCLUSIONS: The present data suggest that TTh in severely obese, hypogonadal individuals induces metabolically healthier preadipocytes, improving insulin sensitivity, mitochondrial functioning and lipid handling. A potentially protective role for testosterone on the progression of NAFLD, improving hepatic steatosis and reducing intrahepatic triglyceride content, was also envisaged. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02248467, September 25th 2014.


Subject(s)
Hypogonadism , Intra-Abdominal Fat , Lipid Metabolism/drug effects , Liver , Non-alcoholic Fatty Liver Disease , Obesity , Testosterone , Adult , Biopsy/methods , Cross-Sectional Studies , Humans , Hypogonadism/diagnosis , Hypogonadism/drug therapy , Hypogonadism/epidemiology , Insulin Resistance , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Italy/epidemiology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/diagnosis , Obesity/drug therapy , Obesity/metabolism , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Testosterone/administration & dosage , Testosterone/pharmacokinetics , Treatment Outcome
8.
Naunyn Schmiedebergs Arch Pharmacol ; 394(4): 751-761, 2021 04.
Article in English | MEDLINE | ID: mdl-33159802

ABSTRACT

Glomeruli and renal tubule injury in chronic kidney disease (CKD) is reported to involve induction of macrophage activation through the CCL2/CCR2 axis. The effects of inhibitors of the CCL2/CCR2 axis, such as anti-CCL2 antibody and CCR2 antagonist, on kidney function in animal models or humans with kidney dysfunction have been demonstrated. The N-terminal glutamine on immature CCL2 is replaced with pyroglutamate (pE) by glutaminyl cyclase (QC) and isoQC. pE-CCL2 is stable and resistant to peptidases. We hypothesized that inhibiting QC/isoQC activity would lead to the degradation of CCL2, thereby ameliorating CKD and reducing kidney inflammation. To test this hypothesis, we investigated the renoprotective properties of the QC/isoQC inhibitor PQ529 in anti-glomerular basement membrane (GBM) antibody-induced glomerulonephritis Wistar Kyoto (WKY) rats. Three-week repeated administration of PQ529 (30 and 100 mg/kg, twice daily) significantly reduced the serum and urine CCL2 and urinary protein excretion in a dose-dependent manner. Correlations between the urinary protein level and serum or urinary CCL2 levels were confirmed in tested animals. Repeated administration of PQ529 significantly reduced the expression of CD68, a macrophage marker, in the kidney cortex and mononuclear infiltration into the tubulointerstitium. In addition, decreased levels of urinary KIM-1, ß2 microglobulin, and clusterin were detected, suggesting the inhibition of inflammation in both the proximal and distal tubules. These results suggest that PQ529 suppresses the progression of inflammation-induced renal dysfunction by inhibiting the CCL2/CCR2 axis. Inhibition of QC/isoQC may thus be a viable alternative therapeutic approach for treating glomerulonephritis and CKD patients.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Benzimidazoles/therapeutic use , Glomerulonephritis/drug therapy , Imidazolines/therapeutic use , Protective Agents/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Cell Adhesion Molecules/urine , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/blood , Chemokine CCL2/metabolism , Chemokine CCL2/urine , Clusterin/urine , Glomerulonephritis/blood , Glomerulonephritis/metabolism , Glomerulonephritis/urine , Imidazolines/pharmacokinetics , Imidazolines/pharmacology , Interferon-gamma/metabolism , Kidney/drug effects , Kidney/metabolism , Male , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , Rats, Inbred WKY , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/urine , beta 2-Microglobulin/urine
9.
Acta Histochem ; 122(8): 151632, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33128988

ABSTRACT

BACKGROUND: Testicular hyperthermia can have negative effects on male fertility. Despite reported therapeutic benefits of curcumin, several factors often limit its application such as low water solubility and instable structure. Curcumin-loaded superparamagnetic iron oxide nanoparticles (SPIONs) were designed to solve its limitation of use. In the present study, we evaluated the effect of curcumin-loaded SPIONs on transient testicular hyperthermia in mouse. MATERIALS AND METHOD: A total of 18 adult male NMRI mice were divided into three groups (n = 6): I. Controls (Cont), II. Scrotal hyperthermia (Hyp), III. Scrotal hyperthermia + curcumin-loaded iron particles (240 µL) (Hyp + Cur). After seventy days, the animals were sacrificed and used for further molecular and stereological evaluations. RESULTS: Sperm count, motility and viability significantly decreased in group hyp as compared to cont group. Furthermore, Sperm DNA fragmentation and cell apoptosis in testes increased remarkably in group hyp, compared with group cont. Stereological study showed a reduction in number of spermatogenic and Leydig cells, as well as reduced weight and volume of testes in hyp group. Degenerative appearance of testes exposed to hyperthermia was also observed. In addition, higher mRNA expression of inflammatory cytokines (IL1-α, IL6, and TNF-α) was detected in group hyp compared to cont group. However, curcumin-loaded SPIONs alleviated all of the pathologic changes in the Hyp + Cur group compared to the hyp group. CONCLUSION: Here, we used nanoparticle form of curcumin in testicular hyperthermia model and showed its ameliorating effects on testes damages caused by heat stress, which can be an appropriate method to overcome the problems that limit curcumin application in cases with increased intra testicular temperature.


Subject(s)
Antioxidants/pharmacology , Curcumin/pharmacology , Drug Carriers , Hyperthermia/drug therapy , Magnetic Iron Oxide Nanoparticles/administration & dosage , Protective Agents/pharmacology , Animals , Antioxidants/pharmacokinetics , Cell Survival/drug effects , Curcumin/pharmacokinetics , DNA Fragmentation/drug effects , Gene Expression , Heat-Shock Response/drug effects , Hyperthermia/metabolism , Hyperthermia/pathology , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Leydig Cells/drug effects , Leydig Cells/metabolism , Leydig Cells/pathology , Male , Mice , Oxidative Stress/drug effects , Protective Agents/pharmacokinetics , Scrotum/drug effects , Scrotum/metabolism , Scrotum/pathology , Sperm Count , Sperm Motility/drug effects , Spermatogenesis/drug effects , Spermatogenesis/genetics , Spermatozoa/drug effects , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/drug effects , Testis/metabolism , Testis/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Reprod Toxicol ; 96: 349-358, 2020 09.
Article in English | MEDLINE | ID: mdl-32800787

ABSTRACT

Despite the Cox inhibitory anti-inflammatory and antipyretic effects of most widely used non-steroidal anti-inflammatory drugs (NSAIDs), such as Ibuprofen, their chronic use is associated with a plethora of patho-physiological insults. One such toxic effect on testicular tissues is not well studied and the underlying molecular mechanisms remain unexplored. Thus, the current study is designed to evaluate the antioxidant properties of essential trace element selenium (Se) to ameliorative Ibuprofen associated testicular toxic effects. Adult male Wistar rats were divided into 3 groups and fed on diets containing different concentrations of sodium selenite, viz. 0.01 mg/kg (Se- deficient), 0.2 mg/kg (Se-adequate), or 0.5 mg/kg (Se- supplemented) for 8 weeks. After diet feeding schedule, each group was divided into two subgroups i.e., with or without the treatment of Ibuprofen (120 mg/kg Bw). The protective effect of Se was evaluated by measuring testicular Se and selenoproteins status, spermatogenic markers, histopathology and testicular redox status. Ibuprofen diminished seminal volume, sperm count, sperm motility, which correlated well increased testicular reactive oxygen species. Se deficiency exacerbated these detrimental effects of ibuprofen by increasing oxidative stress. Alternatively, Se supplementation through antioxidant enzymes mediated protective effects. Se as essential antioxidant selenoproteins ameliorates Ibuprofen induced male reproductive toxicity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Ibuprofen/toxicity , Protective Agents/therapeutic use , Sodium Selenite/therapeutic use , Testis/drug effects , Animals , Glutathione/metabolism , Glutathione Transferase/metabolism , Male , Oxidation-Reduction , Oxidoreductases/metabolism , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , Rats, Wistar , Sodium Selenite/blood , Sodium Selenite/pharmacokinetics , Sodium Selenite/pharmacology , Spermatozoa/drug effects , Testis/metabolism , Testis/pathology
11.
Int J Nanomedicine ; 15: 5217-5226, 2020.
Article in English | MEDLINE | ID: mdl-32801687

ABSTRACT

AIM: Chronic use of oral nonsteroidal anti-inflammatory drugs (NSAIDs) is commonly associated with gastric irritation and gastric ulceration. Therefore, the aim of study was to develop a novel oral drug delivery system with minimum gastric effects and improved dissolution rate for aceclofenac (ACF), a model BCS class-II drug. METHODS: Self-emulsifying drug delivery systems (SEDDS) were formulated to increase the solubility and ultimately the oral bioavailability of ACF. Oleic acid was used as an oil phase, Tween 80 (T80) and Kolliphor EL (KEL) were used as surfactants, whereas, polyethylene glycol 400 (PEG 400) and propylene glycol (PG) were employed as co-surfactants. Optimized formulations (F1, F2, F3 and F4) were analyzed for droplet size, poly dispersity index (PDI), cell viability studies, in vitro dissolution in both simulated gastric fluid and simulated intestinal fluid, ex vivo permeation studies and thermodynamic stability. RESULTS: The optimized formulations showed mean droplet sizes in the range of 111.3 ± 3.2 nm and 470.9 ± 12.52 nm, PDI from 244.6 nm to 389.4 ± 6.51 and zeta-potential from -33 ± 4.86 mV to -38.5 ± 5.15 mV. Cell viability studies support the safety profile of all formulations for oral administration. The in vitro dissolution studies and ex vivo permeation analysis revealed significantly improved drug release ranging from 95.68 ± 0.02% to 98.15 ± 0.71% when compared with control. The thermodynamic stability studies confirmed that all formulations remain active and stable for a longer period. CONCLUSION: In conclusion, development of oral SEDDS might be a promising tool to improve the dissolution of BCS class-II drugs along with significantly reduced exposure to gastric mucosa.


Subject(s)
Diclofenac/analogs & derivatives , Drug Delivery Systems/methods , Emulsions/chemistry , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Biological Availability , Caco-2 Cells , Cell Survival/drug effects , Diclofenac/administration & dosage , Diclofenac/pharmacokinetics , Drug Liberation , Emulsions/administration & dosage , Excipients/chemistry , Humans , Male , Nanostructures/administration & dosage , Nanostructures/chemistry , Polyethylene Glycols/chemistry , Polysorbates/chemistry , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Rats, Sprague-Dawley , Solubility , Surface-Active Agents/chemistry
12.
J Nanobiotechnology ; 18(1): 71, 2020 May 13.
Article in English | MEDLINE | ID: mdl-32404183

ABSTRACT

BACKGROUND: Although many therapeutic strategies for Alzheimer's disease (AD) have been explored, these strategies are seldom used in the clinic. Therefore, AD therapeutic research is still urgently needed. One major challenge in the field of nanotherapeutics is to increase the selective delivery of drugs to a targeted location. Herein, we devised and tested a strategy for delivery of nanoparticles to neurons to inhibit tau aggregation by directly targeting p-tau. RESULTS: Curcumin (CUR) is loaded onto red blood cell (RBC) membrane-coated PLGA particles bearing T807 molecules attached to the RBC membrane surface (T807/RPCNP). With the advantage of the suitable physicochemical properties of the PLGA nanoparticles and the unique biological functions of the RBC membrane, the RPCNP are stabilized and promote sustained CUR release, which provided improved biocompatibility and resulted in long-term presence in the circulation. Under the synergistic effects of T807, T807/RPCNP can not only effectively penetrate the blood-brain barrier (BBB), but they also possess high binding affinity to hyperphosphorylated tau in nerve cells where they inhibit multiple key pathways in tau-associated AD pathogenesis. When CUR was encapsulated, our data also demonstrated that CUR-loaded T807/RPCNP NPs can relieve AD symptoms by reducing p-tau levels and suppressing neuronal-like cells death both in vitro and in vivo. The memory impairment observed in an AD mouse model is significantly improved following systemic administration of CUR-loaded T807/RPCNP NPs. CONCLUSION: Intravenous neuronal tau-targeted T807-modified novel biomimetic nanosystems are a promising clinical candidate for the treatment of AD.


Subject(s)
Alzheimer Disease , Biomimetic Materials , Curcumin , Drug Carriers , Nanoparticles/chemistry , Animals , Apoptosis/drug effects , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Blood-Brain Barrier/metabolism , Cell Line , Curcumin/chemistry , Curcumin/pharmacokinetics , Curcumin/pharmacology , Disease Models, Animal , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Hippocampus/drug effects , Hippocampus/pathology , Humans , Maze Learning/drug effects , Mice , Neurons/metabolism , Protective Agents/chemistry , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , tau Proteins/metabolism
13.
IET Nanobiotechnol ; 14(4): 341-345, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32463025

ABSTRACT

Lycopene (LYC) is known to protect cells from oxidative damage caused by free radicals in human tissues. In the present study, the authors designed a LYC-loaded sialic acid (SA)-conjugated poly(D,L-lactide-co-glycolide) (PLGA) nanoparticle (LYC-NP) to enhance the therapeutic efficacy of LYC in acute kidney injury. The characteristics of the LYC-NPs were defined according to particle size, morphology, and in vitro drug release. The LYC-NPs exhibited a controlled release of LYC over 48 h. Confocal laser scanning microscopy clearly highlighted the targeting potential of SA. Enhanced green fluorescence was observed for the LYC-NPs in H2O2-treated human umbilical vein endothelial cells, indicating enhanced internalisation of NPs. The LYC-NPs showed significantly greater cell viability than H2O2-treated cells. In addition, the LYC-NPs remarkably reduced proinflammatory cytokine levels, attributable mainly to the increased cellular internalisation of the SA-based carrier delivery system. Furthermore, protein levels of caspase-3 and -9 were significantly down-regulated after treatment with the LYC-NPs. Overall, they have demonstrated that SA-conjugated PLGA-NPs containing LYC could be used to treat kidney injury.


Subject(s)
Drug Carriers , Kidney Diseases/chemically induced , Lycopene , Nanoparticles/chemistry , Protective Agents , Cell Survival/drug effects , Cells, Cultured , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/adverse effects , Kidney Diseases/metabolism , Lycopene/chemistry , Lycopene/pharmacokinetics , Lycopene/pharmacology , N-Acetylneuraminic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Protective Agents/chemistry , Protective Agents/pharmacokinetics , Protective Agents/pharmacology
14.
Food Chem ; 322: 126742, 2020 Aug 30.
Article in English | MEDLINE | ID: mdl-32305872

ABSTRACT

Almond hulls, the main by-product of almond production, are considered a valuable source of bioactive phenolic compounds. This study aimed to characterize the phenolic composition, bioavailability of the phenolic-rich extracts from almond hulls (PEAH), and their protective effect on oxidative stressed Caco-2 cells induced by tert-butylhydroperoxide (t-BOOH). The ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS) analysis detected 11 phenolic compounds in the PEAH with high total phenolic content and antioxidant activity. Oxidative Caco-2 cell damage was reduced by PEAH, especially at 5 µg/mL, through scavenging reactive oxygen species (ROS), modulating the cellular endogenous antioxidant system and cell redox at a predictable status. Also, in vitro digestion influenced the phenolic compounds' composition and antioxidant power of PEAH. These results suggested that almond hulls, rich in phenolic compounds, can meliorate the oxidative stressed Caco-2 cells and restore its impaired redox balance, and ultimately improve health benefits.


Subject(s)
Oxidative Stress/drug effects , Phenols/chemistry , Plant Extracts/chemistry , Protective Agents/pharmacology , Prunus dulcis/chemistry , Antioxidants/chemistry , Biological Availability , Caco-2 Cells , Chromatography, High Pressure Liquid , Discriminant Analysis , Half-Life , Humans , Least-Squares Analysis , Mass Spectrometry , Oxidation-Reduction , Phenols/pharmacology , Protective Agents/chemistry , Protective Agents/pharmacokinetics , Prunus dulcis/metabolism , Reactive Oxygen Species/chemistry , tert-Butylhydroperoxide/toxicity
15.
Food Chem Toxicol ; 139: 111248, 2020 May.
Article in English | MEDLINE | ID: mdl-32156568

ABSTRACT

Polyphenols are secondary metabolites widely distributed in many plant foods, such a tea, coffee, chocolate and fruits. The consumption of these compounds is related to the improvement or amelioration of many diseases, including diabetes. Nevertheless, the great barrier to the therapeutic use of polyphenols is the low bioavailability of these compounds once ingested. For that reason, the encapsulation of polyphenols in different matrices may protect them from digestion and improve their release and subsequent absorption to obtain target-specific health effects. Some studies have reported the beneficial effect of encapsulation to increase both bioavailability and bioaccessibility. However, these works have mostly been carried out in vitro and few studies are specifically addressed at improving diabetes. In the current work, an overview of the knowledge related to nanoparticles and their use in the diabetic condition has been reviewed.


Subject(s)
Diabetes Mellitus/therapy , Polyphenols/pharmacokinetics , Animals , Antioxidants/analysis , Antioxidants/pharmacokinetics , Biological Availability , Cacao/chemistry , Coffee/chemistry , Disease Management , Disease Models, Animal , Evidence-Based Medicine , Fruit/chemistry , Humans , Hypoglycemic Agents/analysis , Hypoglycemic Agents/pharmacokinetics , Nanoparticles/chemistry , Polymers/chemistry , Polyphenols/analysis , Protective Agents/analysis , Protective Agents/pharmacokinetics , Tea/chemistry
16.
Adv Ther ; 37(4): 1279-1301, 2020 04.
Article in English | MEDLINE | ID: mdl-32065376

ABSTRACT

Silymarin, an extract from milk thistle seeds, has been used for centuries to treat hepatic conditions. Preclinical data indicate that silymarin can reduce oxidative stress and consequent cytotoxicity, thereby protecting intact liver cells or cells not yet irreversibly damaged. Eurosil 85® is a proprietary formulation developed to maximize the oral bioavailability of silymarin. Most of the clinical research on silymarin has used this formulation. Silymarin acts as a free radical scavenger and modulates enzymes associated with the development of cellular damage, fibrosis and cirrhosis. These hepatoprotective effects were observed in clinical studies in patients with alcoholic or non-alcoholic fatty liver disease, including patients with cirrhosis. In a pooled analysis of trials in patients with cirrhosis, silymarin treatment was associated with a significant reduction in liver-related deaths. Moreover, in patients with diabetes and alcoholic cirrhosis, silymarin was also able to improve glycemic parameters. Patients with drug-induced liver injuries were also successfully treated with silymarin. Silymarin is generally very well tolerated, with a low incidence of adverse events and no treatment-related serious adverse events or deaths reported in clinical trials. For maximum benefit, treatment with silymarin should be initiated as early as possible in patients with fatty liver disease and other distinct liver disease manifestations such as acute liver failure, when the regenerative potential of the liver is still high and when removal of oxidative stress, the cause of cytotoxicity, can achieve the best results.


Subject(s)
Liver Diseases/drug therapy , Protective Agents/pharmacology , Protective Agents/therapeutic use , Silymarin/pharmacology , Silymarin/therapeutic use , Blood Glucose/drug effects , Diabetes Mellitus/physiopathology , Hepatocytes , Humans , Liver Cirrhosis/drug therapy , Liver Diseases, Alcoholic/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Silymarin/administration & dosage , Silymarin/pharmacokinetics
17.
Expert Opin Drug Deliv ; 17(2): 237-253, 2020 02.
Article in English | MEDLINE | ID: mdl-32003249

ABSTRACT

Background: The poor biopharmaceutical properties of thymoquinone (TQ) obstruct its development as a hepatoprotective agent. To surmount the delivery challenges of TQ, phospholipid nanoconstructs (PNCs) were constructed.Method: PNCs were constructed employing microemulsification technique and systematic optimization by three-factor three level Box-Behnken design.Result: Optimized PNC composition exhibited nano size (<100 nm), spherical morphology, within acceptable range of polydispersity index (0.55), high drug entrapment efficiency (>90%), controlled drug release pattern, and neutral surface charge (zeta potential of -0.65 mV). After oral administration of a single dose of PNC, it showed a relative bioavailability of 386.03% vis-à-vis plain TQ suspension. Further, TQ-loaded PNC demonstrated significant enhanced hepato-protective effect vis-à-vis pure TQ suspension and silymarin, as evidenced by reduction in the ALP, ALT, AST, bilirubin, and albumin level and ratified by histopathological analysis.Conclusion: TQ-loaded PNCs can be efficient nano-platforms for the management of hepatic disorders and promising drug delivery systems to enhance oral bioavailability of this hydrophobic molecule.


Subject(s)
Benzoquinones/administration & dosage , Benzoquinones/pharmacokinetics , Nanoparticles/administration & dosage , Phospholipids/administration & dosage , Phospholipids/pharmacokinetics , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Drug Delivery Systems , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases/prevention & control , Nanoparticles/chemistry , Rats, Wistar , Suspensions
18.
Curr Radiopharm ; 12(3): 211-219, 2019.
Article in English | MEDLINE | ID: mdl-31612808

ABSTRACT

BACKGROUND: Nephrotoxicity is a prevalent consequence of cancer treatment using radiotherapy and chemotherapy or their combination. There are two methods; histological and biochemical, to assess the kidney damage caused by toxic agents in animal studies. Although these methods are used for the try-out of renoprotective factors, these methods are invasive and time-consuming, and also, lack the necessary sensitivity for primary diagnosis. Quantitative renal 99mTc-DMSA scintigraphy is a noninvasive, precise and sensitive radionuclide technique which is used to assess the extent of kidney damage, so that the extent of injury to the kidney will be indicated by the renal uptake rate of 99mTc-DMSA in the kidney. In addition, this scintigraphy evaluates the effect of the toxic agents by quantifying the alterations in the biodistribution of the radiopharmaceutical. CONCLUSION: In this review, the recent findings about the renoprotective agents were evaluated and screened with respect to the use of 99mTc-DMSA , which is preclinically and clinically used for animal cases and cancer patients under the treatment by radiotherapy and chemotherapy.


Subject(s)
Kidney Neoplasms/metabolism , Kidney/metabolism , Protective Agents/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Technetium Tc 99m Dimercaptosuccinic Acid/pharmacokinetics , Animals , Humans , Kidney/diagnostic imaging , Kidney/drug effects , Kidney/radiation effects , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/drug therapy , Kidney Neoplasms/radiotherapy , Radionuclide Imaging
19.
Toxicol Appl Pharmacol ; 380: 114704, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31400413

ABSTRACT

Cisplatin (CDDP) is the most commonly used chemotherapeutic drug and has an irreplaceable role in cancer treatment. However, CDDP-induced acute kidney injury (AKI) greatly limits its use. Abundant evidence has confirmed that apoptosis contributes to AKI caused by CDDP administration. The nanoparticle form of selenium, also known as Se@SiO2 nanocomposites (NPs), has been proven to be a potential agent to prevent apoptotic cell death. In this article, we established acute kidney injury models in vivo via a single injection of CDDP and used human kidney 2 (HK-2) cells for experiments in vitro. We demonstrated that NPs can improve CDDP-induced renal dysfunction. In addition, therapy with NPs attenuated apoptosis in cells and kidney tissues treated with CDDP. In terms of mechanism, we discovered that Sirt1, a deacetylase with an important role in CDDP-induced acute kidney injury, was remarkedly increased after NPs pretreatment, and the anti-apoptotic effect of the NPs was markedly abrogated after the inhibition of Sirt1. The results linked the protective effect of NPs on nephrotoxicity with Sirt1, suggesting the potential clinical importance of nanomaterials in alleviating the side effects of chemotherapy.


Subject(s)
Acute Kidney Injury/drug therapy , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Nanospheres/therapeutic use , Protective Agents/therapeutic use , Selenium/therapeutic use , Silicon Dioxide/therapeutic use , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Cell Line , Female , Humans , Interleukin-6/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice, Inbred C57BL , Porosity , Protective Agents/pharmacokinetics , Selenium/pharmacokinetics , Silicon Dioxide/pharmacokinetics , Sirtuin 1/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Eur J Pharm Sci ; 138: 105033, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31382031

ABSTRACT

This study is to investigate pharmacokinetics (PK) and hemorheology (HR) of exogenous phosphocreatine (PCr), a cardio-protective agent, and its active metabolite creatine (Cr), with particular focus on the PK and PD comparison between PCr and Cr. A specific ion-pair reversed-phase HPLC-UV assay was used to simultaneously measure PCr, Cr and ATP concentrations in plasma and red blood cells (RBC) samples of rabbits. PK and HR parameters were calculated based on concentration-time (C-T) curves and effect-time (E-T) curves, respectively, obtained after i.v. dosing. Meanwhile the apparent pharmacological activity ratio (Rapp) and real pharmacological activity ratio (Rreal) of Cr to PCr were calculated. The PCr disappeared from plasma rapidly and in a biphasic manner; plasma PCr was converted to Cr fast and largely with the elimination rate limited metabolite disposition in vivo (Km < K). The i.v. administration of PCr led to a markedly elevated and long-lasting ATP level in RBC. After i.v. administration of preformed Cr, plasma Cr displayed similar elimination kinetics behaviors to that of Cr generated metabolically after i.v. PCr. The Cr could also raise ATP level in RBC, but to less extent than PCr. Approximately 43% of PCr-derived ATP came from Cr-derived ATP in RBC. PCr could significantly reduce whole blood viscosity and RBC osmotic fragility and Cr could do so, but weakly with estimated Rapp of 0.53-0.68 and Rreal of 0.38-0.48. PCr also inhibited platelet aggregation significantly, as opposed to Cr. The PCr-caused improvement of HR is related to the rise in ATP level in RBC. Cr is likely to partially mediate HR effect of PCr.


Subject(s)
Creatine/metabolism , Creatine/pharmacokinetics , Hemorheology/physiology , Phosphocreatine/metabolism , Phosphocreatine/pharmacokinetics , Adenosine Triphosphate/metabolism , Animals , Blood Viscosity/drug effects , Kinetics , Male , Platelet Aggregation/drug effects , Protective Agents/pharmacokinetics , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...