Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Nanomaterials (Basel) ; 12(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36144979

ABSTRACT

Currently, the fast growth and advancement in technologies demands promising supercapacitors, which urgently require a distinctive electrode material with unique structures and excellent electrochemical properties. Herein, binder-free manganese iron sulfide (Mn-Fe-S) nanostructures were deposited directly onto Ni-foam through a facile one-step electrodeposition route in potentiodynamic mode. The deposition cycles were varied to investigate the effect of surface morphologies on Mn-Fe-S. The optimized deposition cycles result in a fragmented porous nanofibrous structure, which was confirmed using Field Emission Scanning Electron Microscopy (FE-SEM). X-ray photoelectron spectroscopy (XPS) confirmed the presence of Mn, Fe, and S elements. The energy dispersive X-ray spectroscopy and elemental mapping revealed a good distribution of Mn, Fe, and S elements across the Ni-foam. The electrochemical performance confirms a high areal capacitance of 795.7 mF cm-2 with a 24 µWh cm-2 energy density calculated at a 2 mA cm-2 current density for porous fragmented nanofiber Mn-Fe-S electrodes. The enhancement in capacitance is due to diffusive-controlled behavior dominating the capacitator, as shown by the charge-storage kinetics. Moreover, the assembled asymmetric coin cell device exhibited superior electrochemical performance with an acceptable cyclic performance of 78.7% for up to 95,000 consecutive cycles.

2.
Biomedicines ; 10(7)2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35885044

ABSTRACT

Obesity represents chronic low-grade inflammation that precipitates type 2 diabetes, cardiovascular disease, and cancer. Berberine (BBR) has been reported to exert anti-obesity and anti-inflammatory benefits. We aimed to demonstrate the underlying immune-modulating mechanisms of anti-obesity effects of BBR. First, we performed in silico study to identify therapeutic targets, describe potential pathways, and simulate BBR docking at M1 and M2 adipose tissue macrophages (ATMs), tumor necrosis factor-α (TNF-α), C-C motif chemokine ligand 2 (CCL2), CCL4, CCL5, and C-X-C motif chemokine receptor 4 (CXCR4). Next, in vivo, we divided 20 C58BL/6 mice into four groups: normal chow, control (high fat diet (HFD)), HFD + BBR 100 mg/kg, and HFD + metformin (MET) 200 mg/kg. We evaluated body weight, organ weight, fat area in tissues, oral glucose and fat tolerance tests, HOMA-IR, serum lipids levels, population changes in ATMs, M1 and M2 subsets, and gene expression of TNF-α, CCL2, CCL3, CCL5, and CXCR4. BBR significantly reduced body weight, adipocyte size, fat deposition in the liver, HOMA-IR, triglycerides, free fatty acids, ATM infiltration, all assessed gene expression, and enhanced the CD206+ M2 ATMs population. In conclusion, BBR treats obesity and its associated metabolic dysfunctions, by modulating ATM recruitment and polarization via chemotaxis inhibition.

3.
Biomedicines ; 10(1)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35052852

ABSTRACT

Obesity causes low-grade inflammation that results in dyslipidemia and insulin resistance. We evaluated the effect of puerarin on obesity and metabolic complications both in silico and in vivo and investigated the underlying immunological mechanisms. Twenty C57BL/6 mice were divided into four groups: normal chow, control (HFD), HFD + puerarin (PUE) 200 mg/kg, and HFD + atorvastatin (ATO) 10 mg/kg groups. We examined bodyweight, oral glucose tolerance test, serum insulin, oral fat tolerance test, serum lipids, and adipocyte size. We also analyzed the percentage of total, M1, and M2 adipose tissue macrophages (ATMs) and the expression of F4/80, tumor necrosis factor-α (TNF-α), C-C motif chemokine ligand 2 (CCL2), CCL4, CCL5, and C-X-C motif chemokine receptor 4. In silico, we identified the treatment-targeted genes of puerarin and simulated molecular docking with puerarin and TNF, M1, and M2 macrophages based on functionally enriched pathways. Puerarin did not significantly change bodyweight but significantly improved fat pad weight, adipocyte size, fat area in the liver, free fatty acids, triglycerides, total cholesterol, and HDL-cholesterol in vivo. In addition, puerarin significantly decreased the ATM population and TNF-α expression. Therefore, puerarin is a potential anti-obesity treatment based on its anti-inflammatory effects in adipose tissue.

SELECTION OF CITATIONS
SEARCH DETAIL
...