Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 137: 112536, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38909495

ABSTRACT

Microglial activation contributes to the neuropathology of Parkinson's disease (PD). Inhibiting M1 while simultaneously boosting M2 microglia activation may therefore be a potential treatment for PD. Apilarnil (API) is a bee product produced from drone larvae. Recent research has demonstrated the protective effects of API on multiple body systems. Nevertheless, its impact on PD or the microglial M1/M2 pathway has not yet been investigated. Thus, we intended to evaluate the dose-dependent effects of API in rotenone (ROT)-induced PD rat model and explore the role of M1/M2 in mediating its effect. Seventy-two Wistar rats were equally grouped as; control, API, ROT, and groups in which API (200, 400, and 800 mg/kg, p.o.) was given simultaneously with ROT (2 mg/kg, s.c.) for 28 days. The high dose of API (800 mg/kg) showed enhanced motor function, higher expression of tyrosine hydroxylase and dopamine levels, less dopamine turnover and α-synuclein expression, and a better histopathological picture when compared to the ROT group and the lower two doses. API's high dose exerted its neuroprotective effects through abridging the M1 microglial activity, illustrated in the reduced expression of miR-155, Iba-1, CD36, CXCL10, and other pro-inflammatory markers' levels. Inversely, API high dose enhanced M2 microglial activity, witnessed in the elevated expression of miR-124, CD206, Ym1, Fizz1, arginase-1, and other anti-inflammatory indices, in comparison to the diseased group. To conclude, our study revealed a novel neuroprotective impact for API against experimentally induced PD, where the high dose showed the highest protection via rebalancing M1/M2 polarization.


Subject(s)
MicroRNAs , Microglia , Neuroprotective Agents , Rats, Wistar , Rotenone , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Male , Rats , Disease Models, Animal , Dopamine/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/genetics
2.
Food Chem Toxicol ; 182: 114119, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37944788

ABSTRACT

BACKGROUND: Doxorubicin (DOX) is a well-known chemotherapeutic agent which causes serious adverse effects due to multiple organ damage, including cardiotoxicity, nephrotoxicity, neurotoxicity, and hepatotoxicity. The mechanism of DOX-induced organ toxicity might be attributed to oxidative stress (OS) and, consequently, activation of inflammatory signaling pathways, apoptosis, and blockage of autophagy. Sophorolipids (SLs) as a glycolipid type of biosurfactants, are natural products that have unique properties and a wide range of applications attributed to their antioxidant and anti-inflammatory properties. AIMS: Production of low-cost SLs from Saccharomyces cerevisiae grown on banana peels and investigating their possible protective effects against DOX-induced hepatotoxicity. MAIN METHODS: The yeast was locally isolated and molecularly identified, then the yielded SLs were characterized by FTIR, 1H NMR and LC-MS/MS spectra. Posteriorly, thirty-two male Wistar rats were randomly divided into four groups; control (oral saline), SLs (200 mg/kg, p.o), DOX (10 mg/kg; i.p.), and SL + DOX (200 mg/kg p.o.,10 mg/kg; i.p., respectively). Liver function tests (LFTs), oxidative stress, inflammatory, apoptosis as well as autophagy markers were investigated. KEY FINDINGS: SLs were produced with a yield of 49.04% and treatment with SLs improved LFTs, enhanced Nrf2 and suppressed NF-κB, IL-6, IL-1ß, p38, caspase 3 and Bax/Bcl2 ratio in addition to promotion of autophagy when compared to DOX group. SIGNIFICANCE: Our results revealed a novel promising protective effect of SLs against DOX-induced hepatotoxicity in rats.


Subject(s)
Chemical and Drug Induced Liver Injury , Musa , Rats , Male , Animals , NF-kappa B/genetics , NF-kappa B/metabolism , Saccharomyces cerevisiae/metabolism , Chromatography, Liquid , Rats, Wistar , Tandem Mass Spectrometry , Doxorubicin/toxicity , Antioxidants/pharmacology , Oxidative Stress , Apoptosis , Cardiotoxicity/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Autophagy
3.
Arch Pharm (Weinheim) ; 356(11): e2300312, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37625018

ABSTRACT

Methotrexate (MTX)-induced hepatotoxicity is a serious adverse effect that may limit its use. Therefore, eligible drugs to ameliorate MTX-induced hepatotoxicity are required. l-Carnitine (LC) is a natural molecule with beneficial metabolic effects and infliximab (INF) is an anti-inflammatory monoclonal antibody against tumor necrosis factor-alpha (TNF-α). Recently, Notch1/Hes-1 signaling was found to play a key role in the pathogenesis of liver injury. However, its role in MTX-induced hepatotoxicity is unclear. This study aimed to evaluate the modulatory effects of LC or INF on MTX-induced hepatotoxicity and to explore the underlying mechanism with emphasis on the Notch1/Hes-1 signaling pathway. Sixty rats were randomized into six groups (n = 10): (1) control (saline); (2) MTX (20 mg/kg MTX, intraperitoneal [ip], once); (3) LC group (500 mg/kg ip, 5 days); (4) INF (7 mg/kg INF ip, once); (5) MTX+LC (20 mg/kg ip, once, 500 mg/kg ip, 5 days, respectively); (6) MTX+INF (20 mg/kg ip, once, 7 mg/kg INF ip, once, respectively). Oxidative stress, inflammatory markers, and Notch1/Hes-1 were investigated. MTX induced the expression of Notch1 and Hes-1 proteins and increased the levels of TNF-α, interleukin (IL)-6, and IL-1ß in the liver. Cotreatment with LC or INF showed apparent antioxidant and anti-inflammatory effects. Interestingly, the downregulation of Notch1 and Hes-1 expression was more prominent in LC cotreatment as compared with INF. In conclusion, LC or INF attenuates MTX-induced hepatotoxicity through modulation of Notch1/Hes-1 signaling. The LC ameliorative effect against MTX-induced hepatotoxicity is significantly better than that of INF. Therefore, LC cotreatment may present a safe and therapeutically effective therapy in alleviating MTX-induced hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury , Methotrexate , Rats , Animals , Methotrexate/adverse effects , Methotrexate/metabolism , Infliximab/pharmacology , Infliximab/metabolism , Tumor Necrosis Factor-alpha/metabolism , Carnitine/pharmacology , Carnitine/metabolism , Structure-Activity Relationship , Oxidative Stress , Liver , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/metabolism , Signal Transduction , Receptor, Notch1/metabolism
4.
Arch Pharm Res ; 46(4): 323-338, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36959348

ABSTRACT

Alcoholic liver disease (ALD) refers to hepatic ailments induced by excessive alcohol intake. The pathogenesis of ALD comprises a complex interplay between various mechanistic pathways, among which inflammation and oxidative stress are key players. Boswellic acids (BAs), found in Boswellia serrata, have shown hepatoprotective effects owing to their antioxidant and anti-inflammatory activities, nevertheless, their therapeutic potential against ALD has not been previously investigated. Hence, this study was performed to depict the possible protective effect of BAs and detect their underlying mechanism of action in an experimentally-induced ALD mouse model. Male BALB/c mice were equally categorized into six groups: control, BAs-treated, ALD, and ALD that received BAs at three-dose levels (125, 250, and 500 mg/kg) by oral gavage for 14 days. Results showed that the high dose of BAs had the most protective impact against ALD according to histopathology examination, blood alcohol concentration (BAC), and liver function enzymes. Mechanistic investigations revealed that BAs (500 mg/kg) caused a significant decrease in cytochrome P450 2E1(CYP2E1), nicotine adenine dinucleotide phosphate oxidase (NOX) 1/2/4, p38 mitogen-activated protein kinase (MAPK), and sterol regulatory element-binding protein-1c (SREBP-1c) levels, and the expression of miR-155, yet increased peroxisome proliferator-activated receptor alpha (PPARα) levels. This led to an improvement in lipid profile and reduced hepatic inflammation, oxidative stress, and apoptosis indices. In summary, our study concludes that BAs can protect against ethanol-induced hepatic injury, via modulating NOX/p38 MAPK/PPARα pathways and miR-155 expression.


Subject(s)
Liver Diseases, Alcoholic , MicroRNAs , Animals , Male , Mice , Blood Alcohol Content , Inflammation/metabolism , Liver , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/prevention & control , Liver Diseases, Alcoholic/etiology , MicroRNAs/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , PPAR alpha/metabolism , PPAR alpha/pharmacology , PPAR alpha/therapeutic use , Signal Transduction
5.
Food Chem Toxicol ; 165: 113146, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35595039

ABSTRACT

Nod-like receptor pyrin domain-1 containing 3 (NLRP3) inflammasome/tumor necrosis factor alpha (TNFα)/nuclear factor kappa B (NFκB) inflammatory pathway is known to be involved in the pathogenesis of ulcerative colitis (UC). Inversely, miRNA-223 can exert counter-regulatory effect on NLRP3 expression. The mulberry tree (Morus macroura) fruit is attaining increased importance for its antioxidant and anti-inflammatory activity in addition to its high safety profile. Accordingly, we attempted to explore the possible protective effect of mulberry fruit extract (MFE) in acetic acid (AA)-induced UC rat model. Phytochemical constituents of MFE were characterized using high performance liquid chromatography coupled to mass spectrometry (HPLC-MS). In the in vivo study, three doses of MFE were orally given for seven days before intra-rectal induction of UC by AA on day eight. Screening study revealed that MFE (300 mg/kg) significantly reduced macroscopic and microscopic UC scores. Biochemically, MFE ameliorated oxidative stress, levels of TNFR1, NLRP3, p-NFκB p65, TNFα, IL-1ß, and IL-18, caspase-1 activity, but enhanced miRNA-223 expression. In conclusion, our study provided a novel protective impact for MFE against UC, in which miRNA-223 and TNFα/NFκB/NLRP3 pathway are involved. These results provide a promising step that might encourage further investigations of MFE as a protective agent in UC patients.


Subject(s)
Colitis, Ulcerative , MicroRNAs , Morus , Acetic Acid/analysis , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/prevention & control , Fruit/chemistry , Humans , Inflammasomes/metabolism , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plant Extracts/analysis , Rats , Tumor Necrosis Factor-alpha/genetics
6.
Sci Rep ; 10(1): 13045, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32747644

ABSTRACT

Cyclophosphamide (CP) is a chemotherapeutic agent that induces oxidative stress causing multiple organ damage. Sacubitril/valsartan, is a combined formulation of neprilysin inhibitor (sacubitril) and angiotensin II receptor blocker (valsartan), that induces the protective effect of brain natriuretic peptide. The aim of the current study is to investigate the prophylactic impacts of sacubitril/valsartan versus valsartan against CP-induced lung toxicity in rats. Rats were assigned randomly into 6 groups; control; received corn oil (2 ml/kg/day; p.o. for 6 days), sacubitril/valsartan (30 mg/kg; p.o. for 6 days), valsartan (15 mg/kg; p.o. for 6 days), CP (200 mg/kg; i.p. on day 5), sacubitril/valsartan + CP (30 mg/kg; p.o. for 6 days, 200 mg/kg; i.p. single dose on day 5, respectively), valsartan + CP (15 mg/kg; p.o. for 6 days, 200 mg/kg; i.p. single dose on day 5, respectively). Both sacubitril/valsartan and valsartan produced a significant decrease in the inflammation and fibrosis markers in the BALF, in comparison with the CP group. Both sacubitril/valsartan and valsartan produced an apparent decrease in the relative genes expression of miR-150-3p and NF-κB, as well as a significant decrease in the relative expression of P38 and ERK1/2 MAPKs and an increase in the relative gene expression of Nrf-2, compared to CP group. Intriguingly, sacubitril/valsartan , showed subtle superiority in almost all investigated parameters, compared to valsartan. In conclusion, sacubitril/valsartan effectively abrogated the CP induced lung inflammation and fibrosis, providing a potential promising protection that could be linked to their ability to inhibit miR-150-3p via inhibition of NF-κB and MAPK signaling pathways.


Subject(s)
Aminobutyrates/therapeutic use , Lung Injury/drug therapy , Lung Injury/genetics , MAP Kinase Signaling System , MicroRNAs/genetics , NF-kappa B/metabolism , Protective Agents/therapeutic use , Tetrazoles/therapeutic use , Aminobutyrates/pharmacology , Animals , Antioxidants/metabolism , Biphenyl Compounds , Bronchoalveolar Lavage Fluid/cytology , Cyclophosphamide , Cytokines/metabolism , Drug Combinations , Gene Expression Regulation/drug effects , Heme Oxygenase-1/metabolism , Inflammation Mediators/metabolism , L-Lactate Dehydrogenase/metabolism , Leukocyte Count , Lung/pathology , Lung Injury/chemically induced , Lung Injury/pathology , MAP Kinase Signaling System/drug effects , MicroRNAs/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Natriuretic Peptide, Brain/metabolism , Phosphorylation/drug effects , Protective Agents/pharmacology , Rats , Tetrazoles/pharmacology , Valsartan
7.
Toxicology ; 437: 152439, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32197949

ABSTRACT

Cyclophosphamide (CP) is widely used as chemotherapy in various cancers; however, testicular atrophy has been encountered as an associated adverse effect. Oxidative stress, enhanced endoplasmic reticulum (ER) stress, and subsequent apoptosis are involved in the molecular mechanisms of CP-induced testicular toxicity. In addition to the cardiovascular benefits of LCZ696 (sacubitril/valsartan (VAL)), neprilysin inhibition was shown to mediate Ca2+ sequestration inside the ER. Furthermore, long noncoding RNA taurine-upregulated gene 1 (lncRNA TUG1) was shown to ameliorate apoptosis in various diseases. This tempted us to investigate the possible benefit of LCZ696 against CP-induced testicular dysfunction in rats through neprilysin inhibition axis, and the downstream apoptotic cascade, with highlighting the impact of lncRNA TUG1 in regulating testicular toxicity. Sixty adult male Wistar rats were randomly allocated as control, LCZ696, VAL, CP, CP + LCZ696, and CP + VAL. Testicular atrophy was induced by single-dose injection of CP (200 mg/kg; i.p.). LCZ696 treated group received LCZ696 (30 mg/kg; p.o.) for 6 days, with CP (200 mg/kg; i.p.) single-dose on day 5. LCZ696 increased lncRNA TUG1 expression, improved sperm characteristics, hormonal profile, testicular function, antioxidant defences, and Bcl-2. The histopathological picture and reduced oxidative and ER stress markers, aligned with declined Bax, caspase-3 and the expression of CHOP, PUMA, Noxa, Bim, and p53, with a subtle superior effect over VAL-treated group. In conclusion, the current study highlights the promising impact of LCZ696 in ameliorating chemotherapy-induced testicular atrophy; yet, further investigation regarding longer duration and different doses of LCZ696 is warranted.


Subject(s)
Aminobutyrates/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Apoptosis/drug effects , Cyclophosphamide/toxicity , Neprilysin/antagonists & inhibitors , Protease Inhibitors/pharmacology , RNA, Long Noncoding/metabolism , Testis/drug effects , Tetrazoles/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Atrophy , Biphenyl Compounds , Drug Combinations , Endoplasmic Reticulum Stress/drug effects , Humans , Male , Neprilysin/metabolism , Oxidative Stress/drug effects , RNA, Long Noncoding/genetics , Rats, Wistar , Signal Transduction , Spermatogenesis/drug effects , Spermatozoa/drug effects , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/enzymology , Testis/pathology , Valsartan
SELECTION OF CITATIONS
SEARCH DETAIL
...