AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (6 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Metal Nanoparticles in Atherosclerosis: Applications and Potential Toxicity

Fatemeh Imanparast1,2Mahmood Doosti2Mohammad Ali Faramarzi1( )
Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, P.O. Box 14155-6451, Tehran 1417614411, Iran
Department of Medical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran 1417614411, Iran
Show Author Information

Abstract

As a chronic inflammatory disease, atherosclerosis is responsible for thousands of deaths worldwide each year, and it imposes massive economic costs on individuals and on society. Because of its high importance, the discovery of sensitive and accurate strategies for imaging, targeted drug delivery, and therapeutic monitoring of this condition is essential. In recent years, continuous research has achieved remarkable successes in the use of nanotechnology in the molecular imaging and treatment of atherosclerosis. Among various nanoparticles — such as metallic, polymeric, and lipid — metallic nanoparticles are being considered due to their unique properties for use in treatment and imaging. It should be taken into consideration that some of the metal nanoparticles themselves can cause adverse biological effects, and these effects should be considered important risk factors in the pathological pathways leading up to atherosclerosis. This review provides a description of the applications and potential toxicity of metal nanoparticles in atherosclerosis.

References

[1]

M. H. Amtenbrink, H. Hofmann, X. Montet, Superparamagnetic nanoparticles - a tool for early diagnostics. Swiss Medical Weekly, 2014, 140: 1-8.

[2]

I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer therapy and diagnosis. Advanced Drug Delivery Reviews, 2012, 64: 24-36.

[3]

I. Cicha, C.D. Garlichs, C, Alexiou, Cardiovascular therapy through nanotechnology. European Journal of Nanomedicine, 2014, 6: 63-87.

[4]

J. Conde, G. Doria, P. Baptista, Noble metal nanoparticles applications in cancer. Journal of Drug Delivery, 2012, 6: 1-12.

[5]

S. Jain, D.G. Hirst, J.M. O’sullivan, Gold nanoparticles as novel agents for cancer therapy. British Journal of Radiology, 2012, 85: 101-113.

[6]

K.J. Moore, F.J. Sheedy, E.A. Fisher, Macrophages in atherosclerosis: a dynamic balance. Nat. Annual Review of Immunology, 2013, 13: 709-721.

[7]

L.G. Spagnoli, E. Bonanno, G. Sangiorgi, et al., Role of inflammation in atherosclerosis. Journal of Nuclear Medicine, 2007, 48: 1800-1815.

[8]

T. Cyrus, P.M. Winte, S.D. Caruthers, et al., Magnetic resonance nanoparticles for cardiovascular molecular imaging and therapy. Expert Review of Cardiovascular Therapy, 2005, 3: 705-715.

[9]

M.E. Kooi, V.C. Cappendijk, K.B.J.M. Cleutjens, et al., Accumulation of ultrasmall superparamagnetic particles of iron oxide in human atherosclerotic plaques can be detected by in vivo magnetic resonance imaging, Circulation, 2003, 107: 2453-2458.

[10]

F.M.E. Segers, B. Adel, I. Bot, et al., Scavenger receptorAI-targeted iron oxide nanoparticles for in vivo MRI detection of atherosclerotic lesions. Arteriosclerosis Thrombosis and Vascular Biology., 2013, 33: 1812-1819.

[11]

R.A. Trivedi, J. U-King-Im, M.J. Graves, et al., In vivo detection of macrophages in human carotid atheroma temporal dependence of ultrasmall superparamagnetic particles of iron oxide-enhanced MRI. Stroke, 2004, 35: 1631-1635.

[12]

C. Tu, T.S.C. Ng, H. Sohi, et al., Receptor-targeted iron oxide nanoparticles for molecular MR Imaging of inflamed atherosclerotic plaques. Biomaterials, 2011, 32: 7209-7216.

[13]

A. Gojova, B. Guo, R.S. Kota, et al., Induction of inflammation in vascular endothelial cells by metal oxide nanoparticles: effect of particle composition. Environmental Health Perspectives, 2007, 115: 403-409.

[14]

F. Benetti, L. Bregoli, I. Olivato, et al., Effects of metal (loid)-based nanomaterials on essential element homeostasis: the central role of nanometallomics for nanotoxicology. Metallomics, 2014, 6: 729-747.

[15]

Z. Chen, H. Meng, G. Xing, et al., Acute toxicological effects of copper nanoparticles in vivo, Toxicology Letters, 2006, 163: 109-120.

[16]

A. Gojova, J.T. Lee, H.S. Jung, et al., Effect of cerium oxide nanoparticles on inflammation in vascular endothelial cells. Inhalation Toxicology, 2009, 21: 123-130.

[17]

M. Mogharabi, M Abdollah, M.A. Faramarzi, Toxicity of nanomaterials; an undermined issue. DARU Journal of Pharmaceutical Sciences, 2014, 22: 59.

[18]

T.A. Gaziano, A. Bitton, S. Anand, et al., Growing epidemic of coronary heart disease in low- and middleincome countries. Current Problems in Cardiology, 2010, 35: 72-115.

[19]

J.C. Laguna, M Alegret. Regulation of gene expression in atherosclerosis: insights from microarray studies in monocytes/macrophages. Pharmacogenomics, 2012, 13, 477-95.

[20]

R.B. Singh, S.A. Mengi, N.S. Dhalla. Pathogenesis of atherosclerosis: a multifactorial process. Clinical & Experimental Cardiology, 2002, 7: 40-53.

[21]

H.A. Hadi, C.S. Carr, J.A. Suwaidi. Endothelial dysfunction: cardiovascular risk factors, therapy, and outcome. Vascular Health and Risk Management, 2005, 1: 183-198.

[22]

K.J. Moore, I, Tabas. The cellular biology of macrophages in atherosclerosis. Cell, 2011, 145: 341-355.

[23]

A. Rudijanto, The role of vascular smooth muscle cells on the pathogenesis of atherosclerosis. Acta Medica Indonesiana, 2007, 39: 86-93.

[24]

A. Nitenberg, Hypertension, endothelial dysfunction and cardiovascular risk. Archives des maladies du coeur et des vaisseaux, 2006, 99: 915-21.

[25]

J.A. Beckman, M.A. Creager, P. Libby. Diabetes and atherosclerosis: epidemiology, pathophysiology, and management. The Journal of the American Medical Association, 2002, 287: 2570-2581.

[26]

L. Papatheodorou, N. Weiss, Vascular oxidant stress and inflammation in hyperhomocysteinemia. Antioxidants & Redox Signaling, 2007, 9: 1941-58.

[27]

J.A. Ambrose, R.S. Barua. The pathophysiology of cigarette smoking and cardiovascular disease: an update. Journal of the American College of Cardiology, 2004, 43: 1731-1737.

[28]

D. Steinberg, J.L. Witztum, History of discovery oxidized low-density lipoprotein and atherosclerosis. Arteriosclerosis Thrombosis and Vascular Biology, 2010, 30: 2311-2316.

[29]

J. Davignon, P. Ganz, Role of endothelial dysfunction in atherosclerosis. Circulation, 2004, 109: 27-32.

[30]

N. Chhabra, Endothelial dysfunction-A predictor of atherosclerosis. Internet Journal of Medical Update, 2009, 4: 33-41.

[31]

L.M. Yung, F.P. Leung, X. Yao, et al., Reactive oxygen species in vascular wall. Cardiovascular & Hematological Disorders-Drug Targets, 2006, 6: 1-19.

[32]

K.D. O’Brien, T.O. McDonald, A. Chait, et al., Neovascular expression of E-selectin, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 in human atherosclerosis and their relation to intimal leukocyte content. Circulation, 1996, 93: 672-682.

[33]

D. Siegel-Axel, H. Langer, S. Lindemann, et al., Role of platelets in atherosclerosis and inflammation. Medizinische Klinik, 2006, 101: 467-475.

[34]

A. Schäfer, J. Bauersachs, Endothelial dysfunction, impaired endogenous platelet inhibition and platelet activation in diabetes and atherosclerosis. Current Vascular Pharmacology, 2008, 6: 52-60.

[35]

A.H. Sprague, R.A. Khalil. Inflammatory cytokines in vascular dysfunction and vascular Disease. Biochemical Pharmacology. 2009, 78: 539-552.

[36]

J.J. Boyle, Macrophage activation in atherosclerosis: pathogenesis and pharmacology of plaque rupture. Current Vascular Pharmacology, 2005, 3: 63-68.

[37]

J. Kunz. Matrix metalloproteinases and atherogenesis in dependence of age. Gerontology, 2007, 53: 63-73.

[38]

K. Takahashi, M. Takeya, N. Sakashita. Multifunctional roles of macrophages in the development and progression of atherosclerosis in humans and experimental animals. Medical Electron Microscopy, 2002, 35: 179-203.

[39]

A.C. Doran, N. Meller, C.A. McNamara, The role of smooth muscle cells in the initiation and early progression of atherosclerosis. Arteriosclerosis Thrombosis and Vascular Biology, 2008, 28: 812-819.

[40]

H.J. Duckers, Biological mechanisms of microvessel formation in advanced atherosclerosis: the big five. Trends in Cardiovascular Medicine, 2013, 23: 153-164.

[41]

V.E. Stoneman, M.R. Bennett. Role of apoptosis in atherosclerosis and its therapeutic implications. Clinical Science, 2004, 107: 343-354.

[42]

I. Tabas, Macrophage apoptosis in atherosclerosis: consequences on plaque progression and the role of endoplasmic reticulum stress. Antioxidants & Redox Signaling, 2009, 11: 2333-2339.

[43]

G. Almer, M. Saba-Lepek, S. Haj-Yahya, et al., Globular domain of adiponectin: promising target molecule for detection of atherosclerotic lesions. Biologics, 2011a, 5: 95-105.

[44]

G. Almer, K. Wernig, M. Saba-Lepek, et al., Adiponectin-coated nanoparticles for enhanced imaging of atherosclerotic plaques. International Journal of Nanomedicine, 2011b, 6: 1279-1290.

[45]

D.J. You, G. Saravanakumar, S. Son, et al., Dextran sulfate-coated superparamagnetic iron oxide nanoparticles as a contrast agent for atherosclerosis imaging. Carbohydrate Polymers, 2014, 101: 1225-1233.

[46]

M.A. Bruckman, K. Jiang, E.J. Simpson, et al., Dualmodal magnetic resonance and fluorescence imaging of atherosclerotic plaques in vivo using VCAM-1 targeted tobacco mosaic virus. Nano Letters, 2014, 14: 1551-1558.

[47]

M. Nahrendorf, F.A. Jaffer, K.A Kelly, et al., Noninvasive vascular cell adhesion molecule-1 imaging identifies inflammatory activation of cells in atherosclerosis. Circulation. 2006, 114, 1504-1511.

[48]

A. Broisat, S. Hernot, J. Toczek, et al., Nanobodies targeting Mouse/human VCAM1 for the nuclear imaging of atherosclerotic lesions. Circulation Research, 2012, 110: 927-937.

[49]

S. Lee, K.G. Birukov, C.E. Romanoski, et al., Role of phospholipid oxidation products in atherosclerosis. Circular Reference, 2012, 111: 778-799.

[50]

T. Saxer, A. Zumbuehl, B. Müller, The use of shear stress for targeted drug delivery. Cardiovascular Research, 2013, 99: 328-333.

[51]

C. Muhlen, A. Fink-Petri, J. Salaklang, et al., Imaging monocytes with iron oxide nanoparticles targeted towards the monocyte integrin MAC-1 (CD11b/CD18) does not result in improved atherosclerotic plaque detection by in vivo MRI. Contrast Media & Molecular Imaging, 2010, 5: 268-275.

[52]

D.P. Cormode, E. Roessl, A. Thran, et al., Atherosclerotic Plaque Composition: Analysis with multicolor CT and targeted gold nanoparticles. Radiology, 2010, 26: 774-782.

[53]

P. Nazari, R. Dowlatabadi-Bazaz, M.R. Mofid, et al., The antimicrobial effects and metabolomic footprinting of carboxyl-capped bismuth nanoparticles against Helicobacter pylori. Applied Biochemistry and Biotechnology., 2014, 172: 570-579.

[54]

S. Ghasemi, M. Heidary, M.A. Faramarzi, et al., Immobilization of lipase on Fe3O4/ZnO core/shell magnetic nanoparticles and catalysis of Michael-type addition to chalcone derivatives. Journal of Molecular Catalysis B: Enzymatic, 2013, 7: 100-108.

[55]

S. Ghasemi, A. Sadighi, M. Heidary, Immobilisation of lipase on the surface of magnetic nanoparticles and non-porous glass beads for regioselective acetylation of prednisolone. IET Nanobiotechnology, 2013, 7: 100-108.

[56]

T.Y. Tang, S.P.S. Howarth, S.R. Miller, et al., The ATHEROMA (atorvastatin therapy: effects on reduction of macrophage activity) study evaluation using ultrasmall superparamagnetic iron oxide-enhanced magnetic resonance imaging in carotid disease. The Journal of the American College of Cardiology, 2009, 53: 2039-2050.

[57]

K. Tsuchiy, N. Nitta, A. Sonod, et al., Evaluation of atherosclerotic lesions using dextran- and mannandextran-coated USPIO: MRI analysis and pathological findings. International Journal of Nanomedicine, 2012, 7: 2271-2280.

[58]

R. Namgung, K. Singha, M.K. Yu, et al., Hybrid superparamagnetic iron oxide nanoparticle-branched polyethylenimine magnetoplexes for gene transfection of vascular endothelial cells. Biomaterials, 2010, 31: 4204-4213.

[59]

J.S. Ma, W.J. Kim, J.J. Kim, et al., Gold nanoparticles attenuate LPS-induced NO production through the inhibition of NF-jB and IFN-b/STAT1 pathways in RAW264.7 cells. Nitric Oxide, 2010, 23: 214-219.

[60]

C. Yuan, W.S. Kerwin, V.L. Yarnykh, et al., MRI of atherosclerosis in clinical trials. NMR in Biomedicine, 2006, 19: 636-654.

[61]

C.L. Korte, H.C. Hansen, A.F.W. Steen, Vascular ultrasound for atherosclerosis imaging. Interface Focus, 2011, 1: 565-575.

[62]

H. Orbay, H. Hong, Y. Zhang, et al., Positron emission tomography imaging of atherosclerosis. Theranostics, 2013, 3: 894-902.

[63]

S. Achenbach, P. Raggi, Imaging of coronary atherosclerosis by computed tomography. European Heart Journal, 2010, 31: 1442-1448.

[64]

L.C. Winkel, H.C. Groen, B.S. Van Thiel, et al., Folate receptor-targeted single-photon emission computed tomography/computed tomography to detect activated macrophages in atherosclerosis: can it distinguish vulnerable from stable atherosclerotic plaques? Molecular Imaging, 2013, 12: 1-5.

[65]

S. Marrache, S. Dhar. Biodegradable synthetic highdensity lipoprotein nanoparticles for atherosclerosis. Proceedings of the National Academy of Sciences, 2013, 110: 9445-9450.

[66]

G. Saravanakumar, K. Kim, J.H. Park, et al., Current status of nanoparticle-based imaging agents for early diagnosis of cancer and atherosclerosis. Journal of Biomedical Nanotechnology, 2009, 5: 20-35.

[67]

C. Psarros, R. Lee, M. Margaritis, et al., Nanomedicine for the prevention, treatment and imaging of atherosclerosis. Maturitas, 2012, 73: 52-60.

[68]

A. Giasuddin, K. Jhuma, M. Haq, Use of gold nanoparticles in diagnostics, surgery and medicine: A Review. Bangladesh Journal of Medical Biochemistry, 2012, 5: 56-60.

[69]

H.L. Karlsson, J. Gustafsson, P. Cronholm, et al., Size-dependent toxicity of metal oxide particles — A comparison between nano- and micrometer size. Toxicology Letters, 2009, 188: 112-118.

[70]

S. Litovsky, M. Madjid, A. Zarrabi, et al., Superparamagnetic iron oxide-based method for quantifying recruitment of monocytes to mouse atherosclerotic lesions in vivo: enhancement by tissue necrosis factor-alpha, interleukin-1beta, and interferongamma. Circulation, 2003, 107: 1545-1549.

[71]

M. Michalska, Visualization of vascular inflammation in the atherosclerotic mouse by ultrasmall superparamagnetic iron oxide vascular cell adhesion molecule-1-specific nanoparticles. Arteriosclerosis, Thrombosis, and Vascular Biology, 2012, 32: 2350-2357.

[72]

M. Sigovan, Rapid-clearance iron nanoparticles for inflammation imaging of atherosclerotic plaque: initial experience in animal model. Radiology, 2009, 252: 401-409.

[73]

H. Li, M.H. El-Dakdouki, D.C. Zhu, et al., Synthesis of β-cyclodextrin conjugated superparamagnetic iron oxide nanoparticles for selective binding and detection of cholesterol crystals. Chemical Communications, 2012, 48: 3385-3387.

[74]

A. Kunzmann, B. Andersson, C. Vogt, Efficient internalization of silica-coated iron oxide nanoparticles of different sizes by primary human macrophages and dendritic cells. Toxicology and Applied Pharmacology, 2011, 253: 81-93.

[75]

A. Bumb, M.W. Brechbiel, Synthesis and characterization of ultra-small superparamagnetic iron oxide nanoparticles thinly coated with silica. Nanotechnology, 2008, 19: 335601-335606.

[76]

C. Burtea, S. Ballet, S. Laurent, et al., Development of a magnetic resonance imaging protocol for the characterization of atherosclerotic plaque by using vascular cell adhesion molecule-1 and apoptosis-targeted ultrasmall superparamagnetic iron oxide derivatives. Arteriosclerosis Thrombosis and Vascular Biology, 2012, 32: e36-e48.

[77]

S. Howarth, Z.Y. Li, R.A. Trivedi, et al., Correlation of macrophage location and plaque stress distribution using USPIO-enhanced MRI in a patient with symptomatic severe carotid stenosis: a new insight into risk stratification. British Journal of Neurosurgery, 2007, 21: 396-398.

[78]

D.K. Kim, M. Mikhaylova, F.H Wang, et al., Starchcoated superparamagnetic nanoparticles as MR contrast agents. Journal of Materials Chemistry, 2003b, 15: 4343-4351.

[79]

G. Li, K. Huang, Y. Jiang, et al., Preparation and characterization of carboxyl functionalization of chitosan derivative magnetic nanoparticles. Biochemical Engineering Journal, 2008, 40: 408-414.

[80]

G. Li, Y. Jiang, K. Huang, et al., Kinetics of adsorption of Saccharomyces cerevisiae mandelated dehydrogenase on magnetic Fe3O4-chitosan nanoparticles. Colloids and Surfaces A, 2008, 320: 11-18.

[81]

D. Olsen, C. Yang, M. Bodo, et al., Recombinant collagen and gelatin for drug delivery. Advanced Drug Delivery Reviews, 2003, 55: 1547-1567.

[82]

B. Gaihre, S. Aryal, M.S. Khil, et al., Encapsulation of Fe3O4 in gelatin nanoparticles: Effect of different parameters on size and stability of the colloidal dispersion. Journal of Microencapsulation, 2008, 25: 21-30.

[83]

S. Mondini, S. Cenedese, G. Marinoni, et al., One-step synthesis and functionalization of hydroxyl-decorated magnetite Nanoparticles. Journal of Colloid and Interface Science, 2008, 322: 173-179.

[84]

S.M. Moghimi, A.C. Hunter, J.C. Murray. Longcirculating and target-specific nanoparticles: theory to practice. Pharmacological Reviews, 2001, 53: 283-318.

[85]

U. Sakulkhu, M. Mahmoudi, L. Maurizi, et al., Protein corona composition of superparamagnetic iron oxide nanoparticles with various physico-chemical properties and coatings. Science Reports, 2014, 4: 5020.

[86]

H.L. Ma, Y.F. Xu, X.R. Qi, et al., Superparamagnetic iron oxide nanoparticles stabilized by alginate: Pharmacokinetics, tissue distribution, and applications in detecting liver cancers. International Journal of Pharmaceutics, 2008, 354: 217-226.

[87]

M. Zhu, B. Wang, Y. Wang, et al., Endothelial dysfunction and inflammation induced by iron oxide nanoparticle exposure: Risk factors for early atherosclerosis. Toxicology Letters, 2011, 203: 162-171.

[88]

S. Wagner, J. Schnorr, A. Ludwig, et al., Contrastenhanced MR imaging of atherosclerosis using citratecoated superparamagnetic iron oxide nanoparticles: calcifying microvesicles as imaging target for plaque characterization. International Journal of Nanomedicine, 2013, 8: 767-779.

[89]

F. Mohammad, T. Arfin. Thermodynamics and electrochemical characterization of core shell type gold‒ coated superparamagnetic iron oxide nanoparticles. Advanced Materials Letters, 2014, 5: 315-324.

[90]

D. Thorek, A. K. Chen, J. Czupryna, et al., Superparamagnetic iron oxide nanoparticle probes for molecular imaging. Annals of Biomedical Engineering, 2006, 34: 23-38.

[91]

M.A. Faramarzi, A. Sadighi, Insights into biogenic and chemical production of inorganic nanomaterials and nanostructures. Advances in Colloid and Interface Science, 2013, 189-190: 1-20.

[92]

J. Lodhia, G. Mandarano, N.J. Ferris, et al., Development and use of iron oxide nanoparticles (Part 1): Synthesis of iron oxide nanoparticles for MRI. The Biomedical Imaging and Intervention Journal, 2010, 6: e12.

[93]

R. Thomas, I. Park, Y.Y. Jeong, Magnetic iron oxide nanoparticles for multimodal imaging and therapy of cancer. International Journal of Molecular Sciences, 2013, 14: 15910-15930.

[94]

S. Metz, G. Bonaterra, M. Rudelius, et al., Capacity of human monocytes to phagocytose approved iron oxide MR contrast agents in vitro. European Radiology, 2004, 14: 1851-1858.

[95]

M. Settles, M. Etzrodt, K. Kosanke, et al., Different capacity of monocyte subsets to phagocytose iron-oxide nanoparticles. PLoS One, 2011, 6: e25197.

[96]

S.G. Ruehm, C. Corot, P. Vogt, Magnetic resonance imaging of atherosclerotic plaque with ultrasmall superparamagnetic particles of iron oxide in hyperlipidemic rabbits. Circulation, 2001, 103: 415-422.

[97]

K. Morishige, D.F. Kacher, P. Libby, et al., Highresolution magnetic resonance imaging enhanced with superparamagnetic nanoparticles measures macrophage burden in atherosclerosis, Circulation, 2010, 122: 1707-1715.

[98]

M. Sigovan, L. Boussel, A. Sulaiman, et al., Rapidclearance iron nanoparticles for inflammation imaging of atherosclerotic plaque: initial experience in animal model. Radiology, 2009, 252: 401-409.

[99]

J.B. Morris, A.R. Olzinski, R.E. Bernard, et al., MAPK inhibition reduces aortic ultrasmall superparamagnetic iron oxide uptake in a mouse model of atherosclerosis MRI assessment. Arteriosclerosis Thrombosis and Vascular Biology, 2008, 28: 265-271.

[100]

V.V. Kunjathoor, M. Febbraio, E.A. Podrez, et al., cavenger receptors class A-Ⅰ/Ⅱ and CD36 are the principal receptors responsible for the uptake of modified low density lipoprotein leading to lipid loading in macrophages. The Journal of Biological Chemistry, 2002, 277: 49982-49988.

[101]

K.J. Moore, M.W. Freeman, Scavenger receptors in atherosclerosis: beyond lipid uptake. Arterioscler. Arteriosclerosis Thrombosis and Vascular Biology, 2006, 26: 1702-1711.

[102]

B.R. Jarrett, C. Correa, K.L. Ma, et al., In vivo mapping of vascular inflammation using multimodal imaging. PLoS One, 2010, 5: e13254.

[103]

X. Li, M.C. Kramer, C.M. van der Loos, et al., A pattern of disperse plaque microcalcifications identifies a subset of plaques with high inflammatory burden in patients with acute myocardial infarction. Atherosclerosis, 2011, 218: 83-89.

[104]

M. Aikawa, P. Libby. The vulnerable atherosclerotic plaque: pathogenesis and therapeutic approach. Cardiovascular Pathology, 2004, 13: 125-138.

[105]

R. Virmani, A.P. Burke, A. Farb, et al., Pathology of the vulnerable plaque. The Journal of the American College of Cardiology, 2006, 47: C13-C18.

[106]

A.A. Alsheikh-Ali, G.D. Kitsios, E.M. Balk, et al., The vulnerable atherosclerotic plaque: scope of the literature. Annals of Internal Medicine, 2010, 153: 387-395.

[107]

F. Scherer, M. Anton,U. Schillinger, et al., Magnetofection: enhancing and targeting gene delivery by magnetic force in vitro and in vivo. Gene Therapy, 2003, 9: 102-109.

[108]

C. Plank, U. Schillinger, F. Scherer, et al., The magnetofection method: using magnetic force to enhance gene delivery. The Journal of Biological Chemistry, 2003, 384: 737-747.

[109]

Y.S. Kim, Y. Ahn, M.H. Hong, et al., Curcumin attenuates inflammatory responses of TNF-alpha-stimulated human endothelial cells. The Journal of Cardiovascular Pharmacology, 2007, 50: 41-49.

[110]

W.J. Rogers, P. Basu. Factors regulating macrophage endocytosis of nanoparticles: implications for targeted magnetic resonance plaque imaging. Atherosclerosis, 2005, 178: 67-73.

[111]

I. Raynal, P. Prigent, S. Peyramaure, et al., Macrophage endocytosis of superparamagnetic iron oxide nanoparticles mechanisms and comparison of ferumoxides and ferumoxtran-10. Investigative Radiology, 2004, 39: 56-63.

[112]

P. Schuler, D. Assefa, J. Ylänne, et al., Adhesion of monocytes to medical steel as used for vascular stents is mediated by the integrin receptor Mac-1 (CD11b/CD18; alphaM beta2) and can be inhibited by semiconductor coating. Cell Communication and Adhesion, 2003, 10: 17-26.

[113]

C. Muhlen, D. Elverfeldt, N. Bassler, et al., Superparamagnetic iron oxide binding and uptake as imaged by magnetic resonance is mediated by the integrin receptor Mac-1 (CD11b/CD18): Implications on imaging of atherosclerotic plaques. Atherosclerosis, 2007, 193: 102-111.

[114]

D. Pissuwan, T. Niidome, M.B. Cortie, The forthcoming applications of gold nanoparticles in drug and gene delivery systems. Journal of Controlled Release, 2009, 149: 65-71.

[115]

P.M. Tiwari, K. Vig, V.A. Dennis, et al., Functionalized gold nanoparticles and their biomedical applications. Nanomaterials, 2011, 1: 31-63.

[116]

P. Ghosh, G. Han, M. De, et al., Gold nanoparticles in delivery Applications. Advanced Drug Delivery Reviews, 2008, 60: 1307-1315.

[117]

E.E. Connor, J. Mwamuka, A. Gole, et al., Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicity. Small, 2005, 1: 325-327.

[118]

M.A. Faramarzi, H. Forootanfar, Biosynthesis and characterization of gold nanoparticles produced by laccase from Paraconiothyrium variabile. Colloids and Surfaces B: Biointerfaces, 2011, 87: 23-27.

[119]

M. Moshfegh, H. Frorootanfar, B. Zare, et al., Biological synthesis of Au, Ag and Au-Ag bimetallic nanoparticles by alpha-amylase. Digest Journal of Nanomaterials and Biostructures, 2011, 6: 1419-1426

[120]

E.K. Lim, E. Jang, J. Kim, et al., Self-fabricated dextrancoated gold nanoparticles using pyrenyl dextran as a reducible stabilizer and their application as CT imaging agents for atherosclerosis. Journal of Materials Chemistry, 2012, 22; 17518-17524.

[121]

A. Kharlamov, J. Gabinsky, A.N. Perrish, et al., Plasmonic nanophotothermal and stem cell therapy of atherosclerotic plaque as the novel tool for angioplasty and artery remodeling. Rejuvenation Research, 2012, 15: 222-230.

[122]

I.L. Maksimova, G.G. Akchurin, B.N. Khlebtsov, et al., Near- infrared laser photothermal therapy of cancer by using gold nanoparticles: computer simulations and experiment. Medical Laser Application, 2007, 22: 199-206.

[123]

N. Khlebtsov, G. Akchurin, B. Khlebtsov, et al., Laser induced destruction of gold NPs: new weapons in the cellkilling arsenal. SPIE, 2008, 9: 1-3.

[124]

A.N. Kharlamov, A.E. Tyurnina, V.S. Veselova, et al., Plasmonics for treatment of atherosclerosis: results of NANOM-FIM trial. Journal of Nanomedicine & Nanotechnology, 2013, 4: 1-13.

[125]

B. Wang, E. Yantsen, T. Larson, et al., Plasmonic intravascular photoacoustic imaging for detection of macrophages in atherosclerotic plaques. Nano Letters, 2009, 9: 2212-2217.

[126]

A.M. Alkilany, L.B. Thompson, S.P. Boulos, et al., Gold nanorods: their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Advanced Drug Delivery Reviews, 2012, 64: 190-199.

[127]

Z.W.J. Zhang, C. Chen, Gold nanorods based platforms for light-mediated theranostics. Theranostics, 2013, 3: 223-238.

[128]

D. Yeager, Y. Chen, S. Litovsky, et al., Intravascular photoacoustics for image-guidance and temperature monitoring during plasmonic photothermal therapy of atherosclerotic plaques: A feasibility study. Theranostics, 2014, 4: 36-46.

[129]

A.N. Kharlamov, J.L. Gabinsky, Plasmonic photothermic and stem cell therapy of atherosclerotic plaque as a novel nanotool for angioplasty and artery remodeling. Rejuvenation Research, 2012, 15: 222-230.

[130]

T. Taufikurohmah, I.G.M. Sanjaya, A. Baktir, et al., Activity test of nanogold for reduction of free radicals, a pre-assessment utilization nanogold in pharmaceutical as medicines and cosmetics. Materials Science and Engineering: B, 2012, 2: 611-616.

[131]

H.T. Chung, H.O. Pae, B.M. Choi, et al., Nitric oxide as a bioregulator of apoptosis. Biochemical and Biophysical Research Communications, 2001, 282: 1075-1079.

[132]

I. Iavicoli, L. Fontana, V. Leso, et al., The effects of nanomaterials as endocrine disruptors. International Journal of Molecular Sciences, 2013, 14: 16732-16801.

[133]

S. BarathManiKanth, K. Kalishwaralal, M. Sriram, et al., Anti-oxidant effect of gold nanoparticles restrains hyperglycemic conditions in diabetic mice. Journal of Nanobiotechnology, 2010, 8: 16.

[134]

J. Banerjee, N.R. T, Biosynthesis of silver nanoparticles from Syzygium cumini (L.) Seed extract and evaluation of their in vitro antioxidant activities. Digest Journal of Nanomaterials and Biostructures, 2011, 6: 961-968.

[135]

K. Bhol, P. Schechter, Effects of nanocrystalline silver (NPI 32101) in a rat model of ulcerative colitis. Digestive diseases and sciences, 2007, 52: 2732-2742.

[136]

P.L. Nadworny, B.K. Landry, J. Wang, et al., Does nanocrystalline silver have a transferable effect? Wound Repair and Regeneration, 2010, 18: 254-265.

[137]

K.K.Y. Wong, S.O.F. Cheung, L. Huang, et al., Further evidence of the anti-inflammatory effects of silver nanoparticle. Medicinal Chemistry Communications, 2009, 4: 1129-1135.

[138]

H. Brandl, S. Lehmann, M.A. Faramarzi, et al., Biomobilization of silver, gold, and platinum from soil waste materials by HCN-forming microorganisms. Hydrometallurgy, 2008, 94: 14-17.

[139]

K. Kalishwaralal, S. Barathmanikanth, S.R. Pandian, et al., Silver nano - a trove for retinal therapies. Journal of Controlled Release, 2010, 145: 76-90.

[140]

H.H. Lara, N.V. Ayala-Nunez, L. Ixtepan-Turrent, et al., Mode of antiviral action of silver nanoparticles against HIV-1. Journal of Nanobiotechnology, 2010, 8: 1-10.

[141]

S. Gurunathan, K.J. Lee, K. Kalishwaralal, et al., Antiangiogenic properties of silver nanoparticles. Biomaterials, 2009, 30: 6341-6350.

[142]

G.S. Kang, P.A. Gillespie, A. Gunnison, et al., Longterm inhalation exposure to nickel nanoparticles exacerbated atherosclerosis in a susceptible mouse model. Environmental Health Perspectives, 2011, 119: 176-181.

[143]

M.M. Kemp, A. Kumar, S. Mousa, et al., Gold and silver nanoparticles conjugated with heparin derivative possess anti-angiogenesis properties. Nanotechnology, 2009, 20: 455104-455111.

[144]

S. Sheikpranbabu, K. Kalishwaralal, D. Venkataraman, et al., Silver nanoparticles inhibit VEGF-and IL-1β-induced vascular permeability via Src dependent pathway in porcine retinal endothelial cells. Journal of Nanobiotechnology, 2009, 7: 8.

[145]

W.J. Trickler, S.M. Lantz, R.C. Murdock, et al., Silver nanoparticle induced blood-brain barrier inflammation and increased permeability in primary rat brain microvessel endothelial cells. Toxicological Sciences, 2010, 118: 160-170.

[146]

J. Shi, X. Sun, Y. Lin, et al., Endothelial cell injury and dysfunction induced by silver nanoparticles through oxidative stress via IKK/NF-κB pathways. Biomaterials, 2014, 35: 6657-6666.

[147]

P.J. Polverini, Angiogenesis and wound healing: basic discoveries, clinical implications, and therapeutic opportunities. Endodontic Topics, 2012, 24: 130-145

[148]

A. Hoeben, B. Landuyt, M.S. Highley, et al., Vascular endothelial growth factor and angiogenesis. Pharmacological Reviews, 2004, 56: 549-580.

[149]

R. Khurana, M. Simons, J.F. Martin, et al., Role of angiogenesis in cardiovascular disease a critical appraisal. Circulation, 2005, 112: 1813-1824.

[150]

V.J.D. Krouwer, L.H.P. Hekking, M. Langelaar-Makkinje, et al., Endothelial cell senescence is associated with disrupted cell-cell junctions and increased monolayer permeability. Vascular Cell, 2012, 4: 12.

[151]

S. Sheikpranbabu, K. Kalishwaralal, K.J. Lee. The inhibition of advanced glycation end-productsinduced retinal vascular permeability by silver nanoparticles. Biomaterials. 2010, 31, 2260-2271.

[152]

F. Giacco, M. Brownlee. Oxidative stress and diabetic complications. Circulation Research, 2010, 106: 1449-1458.

[153]

S. Sheikpranbabu, K. Kalishwaralal, K.J. Lee, et al., The inhibition of advanced glycation end-productsinduced retinal vascular permeability by silver nanoparticles. Biomaterials, 2010, 31: 2260-2271.

[154]

H. Rosas-Hernandez, S. Jimenez-Badillo, P.P. MartinezCuevas, et al., Effects of 45 nm silver nanoparticles on coronary endothelial cells and isolated rat aortic rings. Toxicology Letters, 2009, 191: 305-313.

[155]

Y.F. Li, Y. Gao, Z. Chaiab, et al., Nanometallomics: an emerging field studying the biological effects of metalrelated nanomaterials. Metallomics, 2014, 6: 220-232.

[156]

K. Peters, R.E. Unger, C.J. Kirkpatrick, et al., Effects of nano-scaled particles on endothelial cell function in vitro: studies on viability, proliferation and inflammation. The Journal of Materials Science: Materials in Medicine, 2004, 15: 321-325.

[157]

H. Yamawaki, N. Iwai. Mechanisms underlying nanosized air-pollution-mediated progression of atherosclerosis: carbon black causes cytotoxic injury/ inflammation and inhibits cell growth in vascular endothelial cells. Circulation Journal, 2006, 70: 129-140.

[158]

M. Yu, Y. Mo, R. Wan, et al., Regulation of plasminogen activator inhibitor-1 expression in endothelial cells with exposure to metal nanoparticles. Toxicology Letters, 2010, 195: 82-89.

[159]

N. Li, M. Hao, R.F. Phalen, et al., Particulate air pollutants and asthma. A paradigm for the role of oxidative stress in PM-induced adverse health effects. Clinical Immunology, 2003a, 109: 250-265.

[160]

N. Li, C. Sioutas, A. Cho, et al., Ultrafine particulate pollutants induce oxidative stress and mitochondrial damage. Environmental Health Perspectives, 2003b, 111: 455-460.

[161]

Y. Suzuki, S. Tada-Oikawa, G. Ichihara, et al., Zinc oxide nanoparticles induce migration and adhesion of monocytes toendothelial cells and accelerate foam cell formation. Toxicology and Applied Pharmacology, 2014, 278: 16-25.

[162]

H. Li, M.H. El-Dakdouki, D.C. Zhu, et al., Synthesis of β-cyclodextrin conjugated superparamagnetic iron oxide nanoparticles for selective binding and detection of cholesterol crystals. Chemical Communications, 2012, 48: 3385-3387.

[163]

T.C. Tsou, S.C. Yeh, F.Y. Tsai, et al. Zinc oxide particles induce inflammatory responses in vascular endothelial cells via NF-κB signaling. Journal of Hazardous Materials, 2010, 183: 1.

[164]

W. Song, J. Zhang, J. Guo, et al., Role of the dissolved zinc ion and reactive oxygen species in cytotoxicity of ZnO nanoparticles. Toxicology Letters, 2010, 199: 389-397.

[165]

C. Kojima, A. Kawakami, T. Takei, et al., Angiotensin converting enzyme inhibitor attenuates monocyte adhesion to vascular endothelium through modulation of intracellular zinc. Journal of Pharmacology and Experimental Therapeutics, 2007, 323: 855-860.

[166]

Z. Bian, T. Tachikawa, P. Zhang, et al., Exhibiting efficient charge separation and unprecedented activity. Journal of the American Chemical Society, 2014, 136: 458-465.

[167]

A. Hu, X. Zhang, D. Luong, et al., Adsorption and photocatalytic degradation kinetics of pharmaceuticals by TiO2 nanowires during water treatment. Waste and Biomass Valorization, 2012, 3: 443-449.

[168]

A. Nemmar, H. Vanbilloen, M.F. Hoylaerts, et al., Passage of intratracheally instilled ultrafine particles from the lung into the systemic circulation in hamster. American Journal of Respiratory and Critical Care Medicine, 2001; 164: 1665-1668.

[169]

L. Mikkelsen, M. Sheykhzade, K.A. Jensen, et al., Modest effect on plaque progression and vasodilatory function in atherosclerosis-prone mice exposed to nanosized TiO2. Particle and Fibre Toxicology, 2011, 8: 32-48.

[170]

A. Montiel-Dávalos, J.L. Ventura-Gallegos, E. AlfaroMoreno, et al., TiO2 nanoparticles induce dysfunction and activation of human endothelial cells. Chemical Research in Toxicology, 2012, 25: 920-930.

[171]

K. Kang, D.H. Lim, I.H. Choi, et al., Vascular tube formation and angiogenesis induced by polyvinylpyrrolidone-coated silver nanoparticles. Toxicology Letters, 2011, 205: 227-234.

[172]

M. Ahamed, Toxic response of nickel nanoparticles in human lung epithelial A549 cells. Toxicology in Vitro, 2011, 25: 930-936.

[173]

W.C. Aird, Endothelial cell heterogeneity. Cold Spring Harbor Perspectives in Medicine, 2014, 2: 1-13.

Nano Biomedicine and Engineering
Pages 111-127
Cite this article:
Imanparast F, Doosti M, Faramarzi MA. Metal Nanoparticles in Atherosclerosis: Applications and Potential Toxicity. Nano Biomedicine and Engineering, 2015, 7(3): 111-127. https://doi.org/10.5101/nbe.v7i3.p111-127

334

Views

11

Downloads

1

Crossref

3

Scopus

Altmetrics

Received: 15 August 2015
Accepted: 26 September 2015
Published: 30 September 2015
© 2015 Fatemeh Imanparast, Mahmood Doosti and Mohammad Ali Faramarzi.

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Return