Generic placeholder image

Current Pharmaceutical Biotechnology

Editor-in-Chief

ISSN (Print): 1389-2010
ISSN (Online): 1873-4316

Research Article

BAO-Ag-NPs as Promising Suppressor of ET-1/ICAM-1/VCAM-1 Signaling Pathway in ISO-induced AMI in Rats

Author(s): Yasser O. Mosaad, Hayam Ateyya, Mohammed A. Hussein, Ahmed M. Moro, Ebtsam A. Abdel-Wahab, Amr A. El-Ella and Zahraa N. Nassar*

Volume 25, Issue 6, 2024

Published on: 11 October, 2023

Page: [772 - 786] Pages: 15

DOI: 10.2174/0113892010256434231010062233

Price: $65

Abstract

Objectives: Acute myocardial infarction (AMI) is the most prevalent cause of myocardial fibrosis and the leading cause of mortality from cardiovascular disease. The goal of this work was to synthesize Balanites aegyptiaca oil-silver nanoparticles (BAO-Ag-NPs) and evaluate their cardioprotective effect against ISO-induced myocardial infarction in rats, as well as their mechanism.

Materials and Methods: BAO was isolated, and the unsaturated fatty acids were estimated. BAO-Ag-NPs was prepared, LD50 was calculated to evaluate its cardioprotective activity against ISO (85 mg/kg)-induced AMI. Different doses of BAO-Ag-NPs (1/50 LD50; 46.6 mg/kg.b.w and 1/20 LD50; 116.5 mg) were received to the rats.

Results: The total fatty acids and unsaturated fatty acids generated by BAO were 909.63 and 653.47 mg/100 g oil, respectively. Oleic acid methyl ester, 9-octadecenoic acid methyl ester, and 9, 12-Octadecadienoic acid methyl ester were the predominant ingredients, with concentrations of 107.6, 243.42, and 256.77 mg/100 g oil, respectively. According to TEM and DLS examinations, BAO-Ag-NPs have a size of 38.20 ± 2.5 nm and a negative zeta potential of -19.82 ± 0.30 mV, respectively. The LD50 of synthesized BAO-Ag-NPs is 2330 mg. On the other hand, BAOAg- NPs reduce myocardial necrosis by lowering increased BNP, cTnI, CK-MB, TC, TG, MDA, MMP2, TGF-β1, PGE2, and IL-6 levels. Furthermore, BAO-Ag-NPs inhibit the expression of ET-1, ICAM-1, and VCAM-1 genes as well as enhance HDL-C, CAT, and GSH levels when compared to the ISO-treated group of rats. Histopathological findings suggested that BAO-Ag- NPs enhance cardiac function by increasing posterior wall thickness in heart tissues.

Conclusion: BAO-Ag-NPs protect against AMI in vivo by regulating inflammation, excessive autophagy, and oxidative stress, as well as lowering apoptosis via suppression of the ET-1, ICAM-1, and VCAM-1 signaling pathways.

Keywords: BAO-Ag-NPs, AMI, CAT, GSH, IL-6, ET-1, ICAM-1, VCAM-1.

Graphical Abstract
[1]
Geng, Z.H.; Huang, L.; Song, M.B.; Song, Y.M. Protective effect of a polysaccharide from Salvia miltiorrhiza on isoproterenol (ISO)-induced myocardial injury in rats. Carbohydr. Polym., 2015, 132, 638-642.
[http://dx.doi.org/10.1016/j.carbpol.2015.06.086] [PMID: 26256391]
[2]
Dhivya, V.; Priya, L.B.; Chirayil, H.T.; Sathiskumar, S.; Huang, C.Y.; Padma, V.V. Piperine modulates isoproterenol induced myocardial ischemia through antioxidant and anti-dyslipidemic effect in male Wistar rats. Biomed. Pharmacother., 2017, 87, 705-713.
[http://dx.doi.org/10.1016/j.biopha.2017.01.002] [PMID: 28088738]
[3]
Abou-Taleb, N.I.; Elblasy, O.A.; Elbesoumy, E.A.; Basuny, H.I.; Elhamadi, E.A.; Nasr eldin, M.S.; Emara, A.A.; Ali, A.A.; Salem, M.A.; Ahmed, F.M.; Hussein, M.A. Mechanism of antiangiogenic and antioxidant activity of newly synthesized CAMBA in ehrlich ascites carcinoma-bearing mice. Asian J. Chem., 2021, 33(10), 2465-2471.
[http://dx.doi.org/10.14233/ajchem.2021.23310]
[4]
Elgizawy, H.A.; Ali, A.A.; Hussein, M.A. Resveratrol: Isolation, and its nanostructured lipid carriers, inhibits cell proliferation, induces cell apoptosis in certain human cell lines carcinoma and exerts protective effect against paraquat-induced hepatotoxicity. J. Med. Food, 2021, 24(1), 89-100.
[http://dx.doi.org/10.1089/jmf.2019.0286] [PMID: 32580673]
[5]
El-gizawy, H.A.E.; Hussein, M.A. Isolation, structure elucidation of ferulic and coumaric acids from fortunella japonica swingle leaves and their structure antioxidant activity relationship. Free Radic. Antioxid., 2016, 7(1), 23-30.
[http://dx.doi.org/10.5530/fra.2017.1.4]
[6]
Boarescu, P.M.; Chirilă, I.; Bulboacă, A.E.; Pârvu, A.; Gheban, D.; Sorana, S.D. Isoproterenol induced myocardial infarction in rats: dose identification. Clujul Med., 2018, 91, S39-S40.
[7]
Wang, N.P.; Wang, Z.F.; Tootle, S.; Philip, T.; Zhao, Z.Q. Curcumin promotes cardiac repair and ameliorates cardiac dysfunction following myocardial infarction. Br. J. Pharmacol., 2012, 167(7), 1550-1562.
[http://dx.doi.org/10.1111/j.1476-5381.2012.02109.x] [PMID: 22823335]
[8]
Hussein, M.A. Synthesis of some novel triazoloquinazolines and triazinoquinazolines and their evaluation for anti-inflammatory activity. Med. Chem. Res., 2012, 21(8), 1876-1886.
[http://dx.doi.org/10.1007/s00044-011-9707-0]
[9]
Abdel Maksoud, H.A.; Elharrif, M.G.; Mahfouz, M.K.; Omnia, M.A.; Abdullah, M.H.; Eltabey, M.E. Biochemical study on occupational inhalation of benzene vapours in petrol station. Respir. Med. Case Rep., 2019, 27, 100836.
[http://dx.doi.org/10.1016/j.rmcr.2019.100836] [PMID: 31008048]
[10]
Hussein, M.A. Anti-obesity, antiatherogenic, anti-diabetic and antioxidant activities of J. montana ethanolic formulation in obese diabetic rats fed high-fat diet. Free Radic. Antioxid., 2011, 1(1), 49-60.
[http://dx.doi.org/10.5530/ax.2011.1.9]
[11]
Ezzat, S.M.; Abdel Motaal, A.; El Awdan, S.A.W. In vitro and in vivo antidiabetic potential of extracts and a furostanol saponin from Balanites aegyptiaca. Pharm. Biol., 2017, 55(1), 1931-1936.
[http://dx.doi.org/10.1080/13880209.2017.1343358] [PMID: 28659002]
[12]
Shafik, N.H.; Shafek, R.E.; Michel, H.N.; Eskander, E.F. Phytochemical study and antihyperglycemic effect of Balanites aegyptiaca kernel extract on alloxan induced diabetic male rat. J. Chem. Pharm. Res., 2016, 8, 128-136.
[13]
Hassan, D.M.; Anigo, K.M.; Umar, I.A.; Alegbejo, J.O. Evaluation of phytoconstituent of Balanites aegyptiaca leaves and fruit-mesocarp extracts. M.O.J. Bioorg. Org. Chem, 2017, 1, 228-232.
[14]
Choudhari, A.S.; Mandave, P.C.; Deshpande, M.; Ranjekar, P.; Prakash, O. Phytochemicals in cancer treatment: From preclinical studies to clinical practice. Front. Pharmacol., 2020, 10, 1614.
[http://dx.doi.org/10.3389/fphar.2019.01614] [PMID: 32116665]
[15]
Khamis, G.; Saleh, A.M.; Habeeb, T.H.; Hozzein, W.N.; Wadaan, M.A.M.; Papenbrock, J.; AbdElgawad, H. Provenance effect on bioactive phytochemicals and nutritional and health benefits of the desert date Balanites aegyptiaca. J. Food Biochem., 2020, 44(6), e13229.
[http://dx.doi.org/10.1111/jfbc.13229] [PMID: 32250478]
[16]
Hussein, M.A.; Abdel‐Gawad, S.M. In vivo hepato-protective properties of purslane extracts on paracetamol-induced liver damage. Malays J. Nutr., 2010, 16(1), 161-70.
[http://dx.doi.org/10.1023/A:1015237612018] [PMID: 12049150]
[17]
Al Ashaal, H.A.; Farghaly, A.A.; Abd El Aziz, M.M.; Ali, M.A. Phytochemical investigation and medicinal evaluation of fixed oil of Balanites aegyptiaca fruits (Balantiaceae). J. Ethnopharmacol., 2010, 127(2), 495-501.
[http://dx.doi.org/10.1016/j.jep.2009.10.007] [PMID: 19833185]
[18]
Hussein, M.A.; Ismail, N.E.; Mohamed, A.H.; Borik, R.M.; Ali, A.A.; Mosaad, Y.O. Plasma Phospholipids: A Promising Simple Biochemical Parameter to Evaluate COVID-19 Infection Severity. Bioinform Biol Insights, 2021, 15, 11779322211055891.
[19]
Hussein, MA. Cardioprotective effects of astaxanthin against isoproterenol-induced cardiotoxicity in rats. J. Nutr. Food Sci., 2014, 5(1), 335.
[http://dx.doi.org/10.4172/2155-9600.1000335]
[20]
Kwon, B.; Lee, H.K.; Querfurth, H.W. Oleate prevents palmitate-induced mitochondrial dysfunction, insulin resistance and inflammatory signaling in neuronal cells. Biochim. Biophys. Acta Mol. Cell Res., 2014, 1843(7), 1402-1413.
[http://dx.doi.org/10.1016/j.bbamcr.2014.04.004] [PMID: 24732014]
[21]
Mohamad, E.A.; Mohamed, Z.N.; Hussein, M.A.; Elneklawi, M.S. GANE can improve lung fibrosis by reducing inflammation via promoting p38MAPK/TGF-β1/NF-κB signaling pathway downregulation. ACS Omega, 2022, 7(3), 3109-3120.
[http://dx.doi.org/10.1021/acsomega.1c06591] [PMID: 35097306]
[22]
Asgharzadeh, F.; Hashemzadeh, A.; Yaghoubi, A.; Avan, A.; Nazari, S.E.; Soleimanpour, S.; Hassanian, S.M.; Ferns, G.A.; Rahmani, F.; Khazaei, M. Therapeutic effects of silver nanoparticle containing sulfasalazine on DSS-induced colitis model. J. Drug Deliv. Sci. Technol., 2021, 61, 102133.
[http://dx.doi.org/10.1016/j.jddst.2020.102133]
[23]
Yang, Y.; Guo, L.; Wang, Z.; Liu, P.; Liu, X.; Ding, J.; Zhou, W. Targeted silver nanoparticles for rheumatoid arthritis therapy via macrophage apoptosis and Re-polarization. Biomaterials, 2021, 264, 120390.
[http://dx.doi.org/10.1016/j.biomaterials.2020.120390] [PMID: 32980634]
[24]
Salvadó, L.; Coll, T.; Gómez-Foix, A.M.; Salmerón, E.; Barroso, E.; Palomer, X.; Vázquez-Carrera, M. Oleate prevents saturatedfatty-acid-induced ER stress, inflammation and insulin resistance in skeletal muscle cells through an AMPK-dependent mechanism. Diabetologia, 2013, 56(6), 1372-1382.
[http://dx.doi.org/10.1007/s00125-013-2867-3] [PMID: 23460021]
[25]
Baimark, Y. Preparation of Surfactant-free Linear and Star-shaped Poly(L-lactide)-b-methoxy Polyethylene Glycol Nanoparticles for Drug Delivery. J. Appl. Sci. (Faisalabad), 2012, 12(3), 263-270.
[http://dx.doi.org/10.3923/jas.2012.263.270]
[26]
Schulte, E.; Weber, K. Schnelle herstellung der fettsaeuremethylester aus fetten mit trimethylsulfoniumhydroxid oder natriummethylat. Fat Sci Technol, 1989, 91, 181-183.
[27]
Yang, G.; Wang, C.; Hong, F.; Yang, X.; Cao, Z. Preparation and characterization of BC/PAM-AgNPs nanocomposites for antibacterial applications. Carbohydr. Polym., 2015, 115, 636-642.
[http://dx.doi.org/10.1016/j.carbpol.2014.09.042] [PMID: 25439942]
[28]
Paula, A.P.; Carmen, L.M.; Antelo, A.; Alvarez, M.; Cagide, E.; Vilariño, N.; Vieytes, M.R.; Botana, L.M. Acute Oral Toxicity of Tetrodotoxin in Mice: Determination of Lethal Dose 50 (LD50) and No Observed Adverse Effect Level (NOAEL). Toxins, 2017, 9, 1-7.
[http://dx.doi.org/10.3390/toxins9030075]
[29]
Boshra, S.A.; Hussein, M.A. Cranberry extract as a supplemented food in treatment of oxidative stress and breast cancer induced by N-Methyl-N-Nitrosourea in female virgin rats. Int. J. Phytomed., 2016, 8, 217-227.
[30]
Fossati, P.; Prencipe, L. Serum triglycerides determined colorimetrically with an enzyme that produces hydrogen peroxide. Clin. Chem., 1982, 28(10), 2077-2080.
[http://dx.doi.org/10.1093/clinchem/28.10.2077] [PMID: 6812986]
[31]
Allain, C.C.; Poon, L.S.; Chan, C.S.G.; Richmond, W.; Fu, P.C. Enzymatic determination of total serum cholesterol. Clin. Chem., 1974, 20(4), 470-475.
[http://dx.doi.org/10.1093/clinchem/20.4.470] [PMID: 4818200]
[32]
Burstein, M.; Scholnick, H.R.; Morfin, R. Rapid method for the isolation of lipoproteins from human serum by precipitation with polyanions. J. Lipid Res., 1970, 11(6), 583-595.
[http://dx.doi.org/10.1016/S0022-2275(20)42943-8] [PMID: 4100998]
[33]
Feng, D.; Ling, W.H.; Duan, R.D. Lycopene suppresses LPS-induced NO and IL-6 production by inhibiting the activation of ERK, p38MAPK, and NF-κB in macrophages. Inflamm. Res., 2010, 59(2), 115-121.
[http://dx.doi.org/10.1007/s00011-009-0077-8] [PMID: 19693648]
[34]
Bancroft, G.D.; Steven, A. Theory and Practice of Histological Technique, 4th ed; Churchill Livingstone: New York, 1983, pp. 99-112.
[35]
Parveen, K.; Banse, V.; Ledwani, L. Green synthesis of nanoparticles: Their advantages and disadvantages. AIP Conf. Proc., 1724, 2016.
[http://dx.doi.org/10.1063/1.49451688]
[36]
Guilger-Casagrande, M.; Lima, R. Synthesis of silver Nanoparticles mediated by fungi: A review. Front. Bioeng. Biotechnol., 2019, 7, 287.
[http://dx.doi.org/10.3389/fbioe.2019.00287] [PMID: 31696113]
[37]
Ansari, M.A. One-pot facile green synthesis of silver nanoparticles using seed extract of phoenix dactylifera and their bactericidal potential against MRSA. In: Evid Based Complement Altern Med; , 2018; pp. 1860280-1860289.
[http://dx.doi.org/10.1155/2018/1860280]
[38]
Borik, R.M.; Hussein, M.A. Synthesis, molecular docking, biological potentials, and structure activity relationship of new quinazoline and quinazoline-4-one derivatives. Asian J. Chem., 2021, 33(2), 423-438.
[http://dx.doi.org/10.14233/ajchem.2021.23036]
[39]
Han, S.H.; Yang, B.S.; Kim, H.J. Effectiveness of aromatherapy massage on abdominal obesity among middle aged women. Taehan Kanho Hakhoe Chi, 2003, 33(6), 839-846.
[http://dx.doi.org/10.4040/jkan.2003.33.6.839] [PMID: 15314402]
[40]
Roy, A.; Bulut, O.; Some, S.; Mandal, A.K.; Yilmaz, M.D. Green synthesis of silver nanoparticles: Biomolecule-nanoparticle organizations targeting antimicrobial activity. RSC Adv. Royal Society of Chemistry, 2019, 9(5), 2673-2702.
[http://dx.doi.org/10.1039/C8RA08982E]
[41]
Zhao, X.; Xia, Y.; Li, Q.; Ma, X.; Quan, F.; Geng, C.; Han, Z. Microwave-assisted synthesis of silver nanoparticles using sodium alginate and their antibacterial activity. Colloids Surf. A Physicochem. Eng. Asp., 2014, 444, 180-188.
[http://dx.doi.org/10.1016/j.colsurfa.2013.12.008]
[42]
Mukherjee, P.; Roy, M.; Mandal, B.P.; Dey, G.K.; Mukherjee, P.K.; Ghatak, J.; Tyagi, A.K.; Kale, S.P. Green synthesis of highly stabilized nanocrystalline silver particles by a non-pathogenic and agriculturally important fungus T. asperellum. Nanotechnology, 2008, 19(7), 075103.
[http://dx.doi.org/10.1088/0957-4484/19/7/075103] [PMID: 21817628]
[43]
Gasmalla, H.B.; Idris, A.M.; Shinger, M.I.; Qin, D.; Shan, D.; Lu, X. Balanites aegyptiaca oil synthesized iron oxide nanoparticles: Characterization and antibacterial activity. J. Biomater. Nanobiotechnol., 2016, 7(3), 154-165.
[http://dx.doi.org/10.4236/jbnb.2016.73016]
[44]
Vlaeminck, B.; Dufour, C.; van Vuuren, A.M.; Cabrita, A.R.J.; Dewhurst, R.J.; Demeyer, D.; Fievez, V. Use of odd and branched-chain fatty acids in rumen contents and milk as a potential microbial marker. J. Dairy Sci., 2005, 88(3), 1031-1042.
[http://dx.doi.org/10.3168/jds.S0022-0302(05)72771-5] [PMID: 15738238]
[45]
Zhao, S.Y.; Lee, D.G.; Kim, C.W.; Cha, H.G.; Kim, Y.H.; Kang, Y.S. Synthesis of magnetic nanoparticles of Fe3O4 and CoFe2O4 and their surface modification by surfactant adsorption. Bull. Korean Chem. Soc., 2006, 27(2), 237-242.
[http://dx.doi.org/10.5012/bkcs.2006.27.2.237]
[46]
Zhang, W.; Shi, X.; Huang, J.; Zhang, Y.; Wu, Z.; Xian, Y. Bacitracin-conjugated superparamagnetic iron oxide nanoparticles: synthesis, characterization and antibacterial activity. ChemPhysChem, 2012, 13(14), 3388-3396.
[http://dx.doi.org/10.1002/cphc.201200161] [PMID: 22753190]
[47]
Brady, S.; Jamalfar, S.; York, M.; Scudamore, C.; Roman, I.; Stamp, C.; Swain, A.; Williams, T.; Griffiths, W.; Patterson, L.; Turton, J. Cardiotoxicity of isoproterenol and levels of serum cardiac troponin I in the Han Wistar rat: a threshold dose response study. Toxicol, 2005, 213, 244-251.
[48]
Zhang, J.; Knapton, A.; Lipshultz, S.E.; Weaver, J.L.; Herman, E.H. Isoproterenol-induced cardiotoxicity in sprague-dawley rats: correlation of reversible and irreversible myocardial injury with release of cardiac troponin T and roles of iNOS in myocardial injury. Toxicol. Pathol., 2008, 36(2), 277-278.
[http://dx.doi.org/10.1177/0192623307313010] [PMID: 18349426]
[49]
York, M.; Scudamore, C.; Brady, S.; Chen, C.; Wilson, S.; Curtis, M.; Evans, G.; Griffiths, W.; Whayman, M.; Williams, T.; Turton, J. Characterization of troponin responses in isoproterenol-induced cardiac injury in the Hanover Wistar rat. Toxicol. Pathol., 2007, 35(4), 606-617.
[http://dx.doi.org/10.1080/01926230701389316] [PMID: 17654401]
[50]
Mikaelian, I.; Coluccio, D.; Morgan, K.T.; Johnson, T.; Ryan, A.L.; Rasmussen, E.; Nicklaus, R.; Kanwal, C.; Hilton, H.; Frank, K.; Fritzky, L.; Wheeldon, E.B. Temporal gene expression profiling indicates early up-regulation of interleukin-6 in isoproterenol-induced myocardial necrosis in rat. Toxicol. Pathol., 2008, 36(2), 256-264.
[http://dx.doi.org/10.1177/0192623307312696] [PMID: 18413786]
[51]
Nakajima, Y.; Yamagishi, T.; Hokari, S.; Nakamura, H. Mechanisms involved in valvuloseptal endocardial cushion formation in early cardiogenesis: Roles of transforming growth factor (TGF)-β and bone morphogenetic protein (BMP). Anat. Rec., 2000, 258(2), 119-127.
[http://dx.doi.org/10.1002/(SICI)1097-0185(20000201)258:2<119::AID-AR1>3.0.CO;2-U] [PMID: 10645959]
[52]
Brady, S.; York, M.; Scudamore, C.; Williams, T.; Griffiths, W.; Turton, J. Cardiac troponin I in isoproterenol-induced cardiac injury in the Hanover Wistar rat: studies on low dose levels and routes of administration. Toxicol. Pathol., 2010, 38(2), 287-291.
[http://dx.doi.org/10.1177/0192623309357948] [PMID: 20100841]
[53]
Morrow, D.A.; Cannon, C.P.; Jesse, R.L.; Newby, L.K.; Ravkilde, J.; Storrow, A.B.; Wu, A.H.B.; Christenson, R.H.; Christenson, R.H.; Apple, F.S.; Cannon, C.P.; Francis, G.; Jesse, R.L.; Morrow, D.A.; Newby, L.K.; Ravkilde, J.; Storrow, A.B.; Tang, W.; Wu, A.H.B. National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines: clinical characteristics and utilization of biochemical markers in acute coronary syndromes. Clin. Chem., 2007, 53(4), 552-574.
[http://dx.doi.org/10.1373/clinchem.2006.084194] [PMID: 17384001]
[54]
Hari Senthil Kumar, S.; Anandan, R.; Devaki, T.; Santhosh Kumar, M. Cardioprotective effects of Picrorrhiza kurroa against isoproterenol-induced myocardial stress in rats. Fitoterapia, 2001, 72(4), 402-405.
[http://dx.doi.org/10.1016/S0367-326X(01)00264-7] [PMID: 11395263]
[55]
Farvin, K.H.S.; Anandan, R.; Sankar, T.V.; Nair, P.G.V. Protective effect of squalene against isoproterenol induced myocardial infarction in rats. J. Clin. Biochem. Nutr., 2005, 37(2), 55-60.
[http://dx.doi.org/10.3164/jcbn.37.55]
[56]
Shirafkan, A.; Marjani, A.; Zaker, F. Serum lipid profiles in acute myocardial infarction patients in Gorgan. Biomed. Res. (Aligarh), 2021, 23, 119-124.
[57]
Murthy, H.N.; Yadav, G.G.; Dewir, Y.H.; Ibrahim, A. Phytochemicals and biological activity of desert date (Balanites aegyptiaca (L.) Delile). Plants, 2020, 10(1), 32.
[http://dx.doi.org/10.3390/plants10010032] [PMID: 33375570]
[58]
Bresson, J.; Flywn, A.; Heinonen, M.; Hulsh, K.; Korhonen, H.; Lagiou, P.; Louk, M. Labeling reference intake value for n-3 and n-6 polyunsaturated fatty acids. European Food Safety Authority, 2000, 1, 1-11.
[59]
Schaefer, E.J.; Asztalos, B.F. Cholesteryl ester transfer protein inhibition, high-density lipoprotein metabolism and heart disease risk reduction. Curr. Opin. Lipidol., 2006, 17(4), 394-398.
[http://dx.doi.org/10.1097/01.mol.0000236364.63840.d8] [PMID: 16832162]
[60]
De Caterina, R.; Madonna, R.; Bertolotto, A.; Schmidt, E.B.; Ma-Donna, R.; Lotto, A.B.; Schmidt, E.B. N-3 fatty acids in the treatment of diabetic patients. Diabetes Care, 2007, 30(4), 1012-1026.
[http://dx.doi.org/10.2337/dc06-1332] [PMID: 17251279]
[61]
Morise, A.; Mourot, J.; Riottot, M.; Weill, P.; Fénart, E.; Hermier, D. Dose effect of alpha-linolenic acid on lipid metabolism in the hamster. Reprod. Nutr. Dev., 2005, 45(4), 405-418.
[http://dx.doi.org/10.1051/rnd:2005037] [PMID: 16045889]
[62]
Pullaiah, C.P.; Nelson, V.K.; Rayapu, S.; G v, N.K.; Kedam, T. Exploring cardioprotective potential of esculetin against isoproterenol induced myocardial toxicity in rats: in vivo and in vitro evidence. BMC Pharmacol. Toxicol., 2021, 22(1), 43.
[http://dx.doi.org/10.1186/s40360-021-00510-0] [PMID: 34266475]
[63]
Gaafar, T.; Attia, W.; Mahmoud, S.; Sabry, D.; Aziz, O.A.; Rasheed, D.; Hamza, H. Cardioprotective effects of wharton jelly derived mesenchymal stem cell transplantation in a rodent model of myocardial injury. Int. J. Stem Cells, 2017, 10(1), 48-59.
[http://dx.doi.org/10.15283/ijsc16063] [PMID: 28446005]
[64]
Garrel, C.; Alessandri, J.M.; Guesnet, P.; Al-Gubory, K.H. Omega-3 fatty acids enhance mitochondrial superoxide dismutase activity in rat organs during post-natal development. Int. J. Biochem. Cell Biol., 2012, 44(1), 123-131.
[http://dx.doi.org/10.1016/j.biocel.2011.10.007] [PMID: 22062949]
[65]
Bedlovičová, Z.; Strapáč, I.; Baláž, M.; Salayová, A. A brief overview on antioxidant activity determination of silver nanoparticles. Molecules, 2020, 25(14), 3191.
[http://dx.doi.org/10.3390/molecules25143191] [PMID: 32668682]
[66]
Ahn, E.Y.; Jin, H.; Park, Y. Green synthesis and biological activities of silver nanoparticles prepared by Carpesium cernuum extract. Arch. Pharm. Res., 2019, 42(10), 926-934.
[http://dx.doi.org/10.1007/s12272-019-01152-x] [PMID: 30972559]
[67]
Ahn, E.Y.; Jin, H.; Park, Y. Assessing the antioxidant, cytotoxic, apoptotic and wound healing properties of silver nanoparticles green-synthesized by plant extracts. Mater. Sci. Eng. C, 2019, 101, 204-216.
[http://dx.doi.org/10.1016/j.msec.2019.03.095] [PMID: 31029313]
[68]
Reddy, N.J.; Nagoor Vali, D.; Rani, M.; Rani, S.S. Evaluation of antioxidant, antibacterial and cytotoxic effects of green synthesized silver nanoparticles by Piper longum fruit. Mater. Sci. Eng. C, 2014, 34, 115-122.
[http://dx.doi.org/10.1016/j.msec.2013.08.039] [PMID: 24268240]
[69]
Alshahrani, S.; Rapoport, R.M.; Soleimani, M. Vascular contractile reactivity in hypotension due to reduced renal reabsorption of Na+ and restricted dietary Na+. Naunyn Schmiedebergs Arch. Pharmacol., 2017, 390(3), 321-326.
[http://dx.doi.org/10.1007/s00210-017-1340-0] [PMID: 28108829]
[70]
Whaley-Connell, A.; Habibi, J.; Rehmer, N.; Ardhanari, S.; Hayden, M.R.; Pulakat, L.; Krueger, C.; Ferrario, C.M.; DeMarco, V.G.; Sowers, J.R. Renin Inhibition and AT1R blockade improve metabolic signaling, oxidant stress and myocardial tissue remodeling. Metabolism, 2013, 62(6), 861-872.
[http://dx.doi.org/10.1016/j.metabol.2012.12.012] [PMID: 23352204]
[71]
Pérez-Martínez, P.I.; Rojas-Espinosa, O.; Hernández-Chávez, V.G.; Arce-Paredes, P.; Estrada-Parra, S. Anti‐inflammatory effect of omega unsaturated fatty acids and dialysable leucocyte extracts on collagen‐induced arthritis in DBA/1 mice. Int. J. Exp. Pathol., 2020, 101(1-2), 55-64.
[http://dx.doi.org/10.1111/iep.12348] [PMID: 32459025]
[72]
El Gizawy, H.A.E.H.; Hussein, M.A.; Abdel-Sattar, E. Biological activities, isolated compounds and HPLC profile of Verbascum nubicum. Pharm. Biol., 2019, 57(1), 485-497.
[http://dx.doi.org/10.1080/13880209.2019.1643378] [PMID: 31401911]
[73]
Mosaad, Y.O.; Hussein, M.A.; Ateyya, H.; Mohamed, A.H.; Ali, A.A.; Ramadan Youssuf, A.; Wink, M.; El-Kholy, A.A. Vanin 1 gene role in modulation of iNOS/MCP-1/TGF-β1 signaling pathway in obese diabetic patients. J. Inflamm. Res., 2022, 15, 6745-6759.
[http://dx.doi.org/10.2147/JIR.S386506] [PMID: 36540060]
[74]
El Gizawy, H.A.; Abo-Salem, H.M.; Ali, A.A.; Hussein, M.A. Phenolic profiling and therapeutic potential of certain isolated compounds from Parkia roxburghii against AChE activity as well as GABA A α5, GSK-3β, and p38α MAP-kinase genes. ACS Omega, 2021, 6(31), 20492-20511.
[http://dx.doi.org/10.1021/acsomega.1c02340] [PMID: 34395996]
[75]
M Soliman, S.; Mosallam, S.; Mamdouh, M.A.; Hussein, M.A.; M Abd El-Halim, S. Design and optimization of cranberry extract loaded bile salt augmented liposomes for targeting of MCP-1/STAT3/VEGF signaling pathway in DMN-intoxicated liver in rats. Drug Deliv., 2022, 29(1), 427-439.
[http://dx.doi.org/10.1080/10717544.2022.2032875] [PMID: 35098843]
[76]
Mostafa, M.M.; Amin, M.M.; Zakaria, M.Y.; Hussein, M.A.; Shamaa, M.M.; Abd El-Halim, S.M. Chitosan surface-modified PLGA nanoparticles loaded with cranberry powder extract as a potential oral delivery platform for targeting colon cancer cells. Pharmaceutics, 2023, 15(2), 606.
[http://dx.doi.org/10.3390/pharmaceutics15020606] [PMID: 36839928]
[77]
Ghorab, M.; Ismail, Z.; Abdala, M. Synthesis and biological activities of some novel triazoloquinazolines and triazinoquinazolines containing benzenesulfonamide moieties. Arzneimittelforschung, 2011, 60(2), 87-95.
[http://dx.doi.org/10.1055/s-0031-1296254] [PMID: 20329657]
[78]
Sibson, N.R.; Blamire, A.M.; Perry, V.H.; Gauldie, J.; Styles, P.; Anthony, D.C. TNF-alpha reduces cerebral blood volume and disrupts tissue homeostasis via an endothelin- and TNFR2-dependent pathway. Brain, 2002, 125(11), 2446-2459.
[http://dx.doi.org/10.1093/brain/awf256] [PMID: 12390971]
[79]
Khaksar, S.; Bigdeli, M.R. Intra-cerebral cannabidiol infusion-induced neuroprotection is partly associated with the TNF-α/TNFR1/NF-кB pathway in transient focal cerebral ischaemia. Brain Inj., 2017, 31(13-14), 1932-1943.
[http://dx.doi.org/10.1080/02699052.2017.1358397] [PMID: 28872345]
[80]
Chisaki, K.; Okuda, Y.; Suzuki, S.; Miyauchi, T.; Soma, M.; Ohkoshi, N.; Sone, H.; Yamada, N.; Nakajima, T. Eicosapentaenoic acid suppresses basal and insulin-stimulated endothelin-1 production in human endothelial cells. Hypertens. Res., 2003, 26(8), 655-661.
[http://dx.doi.org/10.1291/hypres.26.655] [PMID: 14567505]
[81]
Vidanapathirana, A.K.; Thompson, L.C.; Herco, M.; Odom, J.; Sumner, S.J.; Fennell, T.R.; Brown, J.M.; Wingard, C.J. Acute intravenous exposure to silver nanoparticles during pregnancy induces particle size and vehicle dependent changes in vascular tissue contractility in Sprague Dawley rats. Reprod. Toxicol., 2018, 75, 10-22.
[http://dx.doi.org/10.1016/j.reprotox.2017.11.002] [PMID: 29154916]
[82]
Giles, L.V.; Tebbutt, S.J.; Carlsten, C.; Koehle, M.S. Effects of low-intensity and high-intensity cycling with diesel exhaust exposure on soluble P-selectin, E-selectin, I-CAM-1, VCAM-1 and complete blood count. BMJ Open Sport Exerc. Med., 2019, 5(1), e000625.
[http://dx.doi.org/10.1136/bmjsem-2019-000625] [PMID: 31803496]
[83]
El-Faham, A.; Al-Rasheed, H.H.; Sholkamy, E.N.; Osman, S.M.; ALOthman, Z.A. Simple approaches for the synthesis of AgNPs in solution and solid phase using modified methoxypolyethylene glycol and evaluation of their antimicrobial activity. Int. J. Nanomedicine, 2020, 15, 2353-2362.
[http://dx.doi.org/10.2147/IJN.S244678] [PMID: 32308387]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy