1,105
Views
42
CrossRef citations to date
0
Altmetric
Review

Silver Nanoparticles for the Therapy of Tuberculosis

ORCID Icon, , ORCID Icon, , , & show all
Pages 2231-2258 | Published online: 31 Mar 2020

References

  • Thoen CO, LoBue PA, Enarson DA, Kaneene JB, de Kantor IN. Tuberculosis: a re-emerging disease in animals and humans. Vet Ital. 2009;45(1):135–181.20391396
  • De Lorenzo S, Tiberi S. Tuberculosis a re-emerging disease. Intern Emerg Med. 2012;7(S3):185–187. doi:10.1007/s11739-012-0822-9
  • World Health Organization. Global Tuberculosis Report 2015, 20th Ed. World Health Organization; 2015.
  • Schneider E, Moore M, Castro KG. Epidemiology of Tuberculosis in the United States. Clin Chest Med. 2005;26(2):183–195. doi:10.1016/j.ccm.2005.02.00715837104
  • Forrellad MA, Klepp LI, Gioffré A, et al. Virulence factors of the Mycobacterium tuberculosis complex. Virulence. 2013;4(1):3–66. doi:10.4161/viru.2232923076359
  • who.tb.99.260.pdf. Available from: https://www.who.int/docstore/gtb/publications/mdrtb/PDF/who.tb.99.260.pdf. Accessed 1121, 2019.
  • 9789241565714-eng.pdf. Available from: https://apps.who.int/iris/bitstream/handle/10665/329368/9789241565714-eng.pdf. Accessed 1124, 2019..
  • Saravanan M, Niguse S, Abdulkader M, et al. Review on emergence of drug-resistant tuberculosis (MDR & XDR-TB) and its molecular diagnosis in Ethiopia. Microb Pathog. 2018;117:237–242. doi:10.1016/j.micpath.2018.02.04729486274
  • Richter E, Weizenegger M, Rüsch-gerdes S, Niemann S. Evaluation of genotype MTBC assay for differentiation of clinical Mycobacterium tuberculosis complex isolates. J Clin Microbiol. 2003;41(6):2672–2675. doi:10.1128/JCM.41.6.2672-2675.200312791901
  • Thoen CO, Steele JH, Kaneene JB. Zoonotic Tuberculosis: Mycobacterium Bovis and Other Pathogenic Mycobacteria. John Wiley & Sons; 2014.
  • Kiers A, Klarenbeek A, Mendelts B, Van Soolingen D, Koëter G. Transmission of Mycobacterium pinnipedii to humans in a zoo with marine mammals. Int J Tuberc Lung Dis off J Int Union Tuberc Lung Dis. 2008;12(12):1469–1473.
  • Alexander KA, Laver PN, Michel AL, et al. Novel Mycobacterium tuberculosis complex pathogen, M. mungi. Emerg Infect Dis. 2010;16(8):1296–1299. doi:10.3201/eid1608.10031420678329
  • Sathiyavimal S, Vasantharaj S, Bharathi D, et al. Biogenesis of copper oxide nanoparticles (CuONPs) using Sida acuta and their incorporation over cotton fabrics to prevent the pathogenicity of Gram negative and Gram positive bacteria. J Photochem Photobiol B. 2018;188:126–134. doi:10.1016/j.jphotobiol.2018.09.01430267962
  • Shankar PD, Shobana S, Karuppusamy I, et al. A review on the biosynthesis of metallic nanoparticles (gold and silver) using bio-components of microalgae: formation mechanism and applications. Enzyme Microb Technol. 2016;95:28–44. doi:10.1016/j.enzmictec.2016.10.01527866624
  • Silvestry-Rodriguez N, Sicairos-Ruelas EE, Gerba CP, Bright KR. Silver as a Disinfectant In: Ware GW, editor. Reviews of Environmental Contamination and Toxicology. Vol. 191 New York: Springer New York; 2007:23–45. doi:10.1007/978-0-387-69163-3_217708071
  • Paladini F, Pollini M. Antimicrobial silver nanoparticles for wound healing application: progress and future trends. Materials. 2019;12(16):2540. doi:10.3390/ma12162540
  • Shanmuganathan R, Karuppusamy I, Saravanan M, Muthukumar H, Ponnuchamy K, Pugazhendhi VSR. Synthesis of silver nanoparticles and their biomedical applications - a comprehensive review. Current Pharmaceut Design2019. doi: 10.2174/1381612825666190708185506
  • Sakamoto K. The pathology of Mycobacterium tuberculosis infection. Vet Pathol. 2012;49(3):423–439. doi:10.1177/030098581142931322262351
  • Mohanty S, Jena P, Mehta R, et al. Cationic antimicrobial peptides and biogenic silver nanoparticles kill mycobacteria without eliciting DNA damage and cytotoxicity in mouse macrophages. Antimicrob Agents Chemother. 2013;57(8):3688–3698. doi:10.1128/AAC.02475-1223689720
  • Montelongo-Peralta LZ, León-Buitimea A, Palma-Nicolás JP, Gonzalez-Christen J, Morones-Ramírez JR. Antibacterial activity of combinatorial treatments composed of transition-metal/antibiotics against Mycobacterium tuberculosis. Sci Rep. 2019;9(1):1–6. doi:10.1038/s41598-019-42049-530626917
  • Maher D, Raviglione M. Global epidemiology of tuberculosis. Clin Chest Med. 2005;26(2):167–182. doi:10.1016/j.ccm.2005.02.00915837103
  • Seung et al. 2015 Multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis.pdf. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4561400/pdf/cshperspectmed-TUB-a017863.pdf. Accessed 1124, 2019.
  • Seung KJ, Keshavjee S, Rich ML. Multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis. Cold Spring Harb Perspect Med. 2015;5:9. doi:10.1101/cshperspect.a017863
  • World Health Organization 2007 - Global tuberculosis control surveillance, plannin.pdf. Available from: https://apps.who.int/iris/bitstream/handle/10665/144567/9241563141_eng.pdf?sequence=1. Accessed 1124, 2019.
  • Chung-Delgado K, Guillen-Bravo S, Revilla-Montag A, Bernabe-Ortiz A. Mortality among MDR-TB cases: comparison with drug-susceptible tuberculosis and associated factors. PLoS One. 2015;10:3. doi:10/f7b7v6
  • Shin SS, Furin JJ, Alcántara F, Bayona J, Sánchez E, Mitnick CD. Long-term follow-up for multidrug-resistant tuberculosis. Emerg Infect Dis. 2006;12(4):687–688. doi:10.3201/eid1204.04125616704823
  • Bernabé-Ortiz A. Factores asociados a supervivencia en pacientes con tuberculosis en Lima, Perú [Factors associated with survival of patients with tuberculosis in Lima, Peru]. Rev Chil Infectologia Organo of Soc Chil Infectologia. 2008;25(2):104–107. Spanish.
  • Ginsberg AM, Spigelman M. Challenges in tuberculosis drug research and development. Nat Med. 2007;13(3):290–294. doi:10.1038/nm0307-29017342142
  • Takiff H, Guerrero E. Current prospects for the fluoroquinolones as first-line tuberculosis therapy. Antimicrob Agents Chemother. 2011;55(12):5421–5429. doi:10.1128/AAC.00695-1121876059
  • Ramachandran G, Swaminathan S. Safety and tolerability profile of second-line anti-tuberculosis medications. Drug Saf. 2015;38(3):253–269. doi:10.1007/s40264-015-0267-y25676682
  • Marra F, Marra CA, Bruchet N, et al. Adverse drug reactions associated with first-line anti-tuberculosis drug regimens. Available from: https://www.ingentaconnect.com/content/iuatld/ijtld/2007/00000011/00000008/art00007. 8 2007 Accessed 122, 2019.
  • Natarajan S, Subramanian P. Adverse drug reactions to second line anti tuberculosis drugs: a prospective study in Mumbai, India. Eur Respir J. 2013;42(Suppl57).
  • Sotgiu G, Centis R, D’ambrosio L, Migliori GB. Tuberculosis treatment and drug regimens. Cold Spring Harb Perspect Med. 2015;5(5):a017822–a017822. doi:10.1101/cshperspect.a01782225573773
  • Crofton SJ, Chaulet P, Maher D, et al. Guidelines for the Management of Drug-Resistant Tuberculosis. Vol No. WHO/TB/96.210 (Rev. 1). Geneva: World Health Organization; 1997.
  • O’Brien RJ, Spigelman M. New drugs for tuberculosis: current status and future prospects. Clin Chest Med. 2005;26(2):327–340. doi:10.1016/j.ccm.2005.02.01315837114
  • Ernst JD. Macrophage receptors for Mycobacterium tuberculosis. Infect Immun. 1998;66(4):1277–1281. doi:10.1128/IAI.66.4.1277-1281.19989529042
  • Porvaznik I, Solovič I, Mokrý J. Non-tuberculous mycobacteria: classification, diagnostics, and therapy. Adv Exp Med Biol. 2017;944:19–25. doi:10.1007/978-3-319-44488-8_4527826888
  • Azadi D, Motallebirad T, Ghaffari K, Shojaei H. Mycobacteriosis and tuberculosis: laboratory diagnosis. Open Microbiol J. 2018;12:41–58. doi:10.2174/187428580181201004129785215
  • Lesley R, Ramakrishnan L. Insights into early mycobacterial pathogenesis from the zebrafish. Curr Opin Microbiol. 2008;11(3):277–283. doi:10.1016/j.mib.2008.05.01318571973
  • Zachary JF, McGavin MD. Pathologic Basis of Veterinary Disease Expert Consult. Elsevier Health Sciences; 2016.
  • Smith I. Mycobacterium tuberculosis pathogenesis and molecular determinants of virulence. Clin Microbiol Rev. 2003;16(3):463–496. doi:10.1128/CMR.16.3.463-496.200312857778
  • Echeverria-Valencia G, Flores-Villalva S, Espitia CI. Virulence factors and pathogenicity of Mycobacterium In: Ribón W editor. Mycobacterium - Research and Development. InTech; 2018:231–255. doi:10.5772/intechopen.72027
  • Torrelles JB, Sieling PA, Zhang N, et al. Isolation of a distinct Mycobacterium tuberculosis mannose-capped lipoarabinomannan isoform responsible for recognition by CD1b-restricted T cells. Glycobiology. 2012;22(8):1118–1127. doi:10.1093/glycob/cws07822534567
  • Fratti RA, Chua J, Vergne I, Deretic V. Mycobacterium tuberculosis glycosylated phosphatidylinositol causes phagosome maturation arrest. Proc Natl Acad Sci. 2003;100(9):5437–5442. doi:10.1073/pnas.073761310012702770
  • Hmama Z. Quantitative analysis of phagolysosome fusion in intact cells: inhibition by mycobacterial lipoarabinomannan and rescue by an 1,25-dihydroxyvitamin D3-phosphoinositide 3-kinase pathway. J Cell Sci. 2004;117(10):2131–2140. doi:10.1242/jcs.0107215090599
  • Chan J, Xuedong F, Shirley WH, Patrick B, Bloom B. Lipoarabinomannan, a possible virulence factor involved in persistence of Mycobacterium tuberculosis within macrophages. Infect Immun. 1991;59(5):1755–1761. doi:10.1128/IAI.59.5.1755-1761.19911850379
  • Shabaana AK, Kulangara K, Semac I, et al. Mycobacterial lipoarabinomannans modulate cytokine production in human T helper cells by interfering with raft/microdomain signalling. CMLS Cell Mol Life Sci. 2005;62(2):179–187. doi:10.1007/s00018-004-4404-515666089
  • Geijtenbeek TBH, van Vliet SJ, Koppel EA, et al. Mycobacteria target DC-SIGN to suppress dendritic cell function. J Exp Med. 2003;197(1):7–17. doi:10.1084/jem.2002122912515809
  • Dao et al. 2004 Mycobacterium tuberculosis lipomannan induces apoptosis and interleukin-12.pdf. Available from: https://iai.asm.org/content/72/4/2067.full.pdf. Accessed 1122, 2019.
  • Quesniaux VJ, Nicolle DM, Torres D, et al. Toll-Like Receptor 2 (TLR2)-dependent-positive and TLR2-independent-negative regulation of proinflammatory cytokines by mycobacterial lipomannans. J Immunol. 2004;172(7):4425–4434. doi:10.4049/jimmunol.172.7.442515034058
  • Indrigo J. Cord factor trehalose 6,6ʹ-dimycolate (TDM) mediates trafficking events during mycobacterial infection of murine macrophages. Microbiology. 2003;149(8):2049–2059. doi:10.1099/mic.0.26226-012904545
  • Axelrod S, Oschkinat H, Enders J, et al. Delay of phagosome maturation by a mycobacterial lipid is reversed by nitric oxide. Cell Microbiol. 2008;10(7):1530–1545. doi:10.1111/j.1462-5822.2008.01147.x18363878
  • Welsh KJ, Abbott AN, Hwang S-A, et al. A role for tumour necrosis factor-, complement C5 and interleukin-6 in the initiation and development of the mycobacterial cord factor trehalose 6,6ʹ-dimycolate induced granulomatous response. Microbiology. 2008;154(6):1813–1824. doi:10.1099/mic.0.2008/016923-018524936
  • Perez RL, Roman J, Roser S, et al. Cytokine message and protein expression during lung granuloma formation and resolution induced by the mycobacterial cord factor trehalose-6,6′-dimycolate. J Interferon Cytokine Res. 2000;20(9):795–804. doi:10.1089/1079990005015106711032399
  • Laneelle G, Tocanne J-F. Evidence for penetration in liposomes and in mitochondrial membranes of a fluorescent analogue of cord factor. Eur J Biochem. 1980;109(1):177–182. doi:10.1111/j.1432-1033.1980.tb04782.x7408875
  • Kato M. Site II-specific inhibition of mitochondrial oxidative phosphorylation by trehalose-6,6′-dimycolate (cord factor) of Mycobacterium tuberculosis. Arch Biochem Biophys. 1970;140(2):379–390. doi:10.1016/0003-9861(70)90079-24319595
  • Ozeki Y, Kaneda K, Fujiwara N, Morimoto M, Oka S, Yano I. In vivo induction of apoptosis in the thymus by administration of mycobacterial cord factor (trehalose 6,6’-dimycolate).. Infect Immun. 1997;65(5):1793–1799. doi:10.1128/IAI.65.5.1793-1799.19979125563
  • Gilleron M, Ronet C, Mempel M, Monsarrat B, Gachelin G, Puzo G. Acylation state of the phosphatidylinositol mannosides from mycobacterium bovis bacillus calmette guérin and ability to induce granuloma and recruit natural killer T cells. J Biol Chem. 2001;276(37):34896–34904. doi:10.1074/jbc.M10390820011441009
  • Court N, Rose S, Bourigault M-L, et al. Mycobacterial PIMs inhibit host inflammatory responses through CD14-Dependent and CD14-Independent mechanisms. PLoS One. 2011;6(9):e24631. doi:10.1371/journal.pone.002463121949737
  • Cambier CJ, Takaki KK, Larson RP, et al. Mycobacteria manipulate macrophage recruitment through coordinated use of membrane lipids. Nature. 2014;505(7482):218–222. doi:10.1038/nature1279924336213
  • Rousseau C, Winter N, Pivert E, et al. Production of phthiocerol dimycocerosates protects Mycobacterium tuberculosis from the cidal activity of reactive nitrogen intermediates produced by macrophages and modulates the early immune response to infection. Cell Microbiol. 2004;6(3):277–287. doi:10.1046/j.1462-5822.2004.00368.x14764111
  • Astarie-Dequeker C, Le Guyader L, Malaga W, et al. Phthiocerol dimycocerosates of M. tuberculosis participate in macrophage invasion by inducing changes in the organization of plasma membrane lipids. PLoS Pathog. 2009;5(2):e1000289. doi:10.1371/journal.ppat.100028919197369
  • Augenstreich J, Arbues A, Simeone R, et al. ESX-1 and phthiocerol dimycocerosates of Mycobacterium tuberculosis act in concert to cause phagosomal rupture and host cell apoptosis. Cell Microbiol. 2017;19(7):e12726. doi:10.1111/cmi.12726
  • Saint-Joanis B, Demangel C, Jackson M, et al. Inactivation of Rv2525c, a substrate of the Twin Arginine Translocation (Tat) System of Mycobacterium tuberculosis, increases -lactam susceptibility and virulence. J Bacteriol. 2006;188(18):6669–6679. doi:10.1128/JB.00631-0616952959
  • Bhuwan M, Arora N, Sharma A, et al. Interaction of Mycobacterium tuberculosis virulence factor RipA with chaperone MoxR1 is required for transport through the TAT secretion system. mBio. 2016;7(2):e02259–15. doi:10.1128/mBio.02259-1526933057
  • Berthet F. Attenuation of virulence by disruption of the Mycobacterium tuberculosis erp Gene. Science. 1998;282(5389):759–762. doi:10.1126/science.282.5389.7599784137
  • Skjot RLV, Oettinger T, Rosenkrands I, et al. Comparative evaluation of low-molecular-mass proteins from mycobacterium tuberculosis identifies members of the ESAT-6 family as immunodominant T-cell antigens. Infect Immun. 2000;68(1):214–220. doi:10.1128/IAI.68.1.214-220.200010603390
  • Elhay MJ, Oettinger T, Andersen P. Delayed-type hypersensitivity responses to ESAT-6 and MPT64 from Mycobacterium tuberculosis in the Guinea Pig.:3. Infect Immun. 1998;66:3454–3456. doi:10.1128/IAI.66.7.3454-3456.19989632623
  • Ganguly N, Giang PH, Gupta C, et al. Mycobacterium tuberculosis secretory proteins CFP-10, ESAT-6 and the CFP10: ESAT6complex inhibit lipopolysaccharide-induced NF-κB transactivation by downregulation of reactive oxidative species (ROS) production. Immunol Cell Biol. 2008;86(1):98–106. doi:10.1038/sj.icb.710011717909563
  • Pathak SK, Basu S, Basu KK, et al. Direct extracellular interaction between the early secreted antigen ESAT-6 of Mycobacterium tuberculosis and TLR2 inhibits TLR signaling in macrophages. Nat Immunol. 2007;8(6):610–618. doi:10.1038/ni146817486091
  • Derrick SC, Morris SL. The ESAT6 protein of Mycobacterium tuberculosis induces apoptosis of macrophages by activating caspase expression. Cell Microbiol. 2007;9(6):1547–1555. doi:10.1111/j.1462-5822.2007.00892.x17298391
  • Smith J, Manoranjan J, Pan M, et al. Evidence for pore formation in host cell membranes by ESX-1-secreted ESAT-6 and its role in Mycobacterium marinum escape from the vacuole. Infect Immun. 2008;76(12):5478–5487. doi:10.1128/IAI.00614-0818852239
  • Kinhikar AG, Verma I, Chandra D, et al. Potential role for ESAT6 in dissemination of Maf tuberculosis via human lung epithelial cells. Mol Microbiol. 2010;75(1):92–106. doi:10.1111/j.1365-2958.2009.06959.x19906174
  • van der Wel N, Hava D, Houben D, et al. M. tuberculosis and M. leprae translocate from the phagolysosome to the cytosol in myeloid cells. Cell. 2007;129(7):1287–1298. doi:10.1016/j.cell.2007.05.05917604718
  • Reed MB, Domenech P, Manca C, et al. A glycolipid of hypervirulent tuberculosis strains that inhibits the innate immune response. Nature. 2004;431(7004):84–87. doi:10.1038/nature0283715343336
  • Alexander KA, Sanderson CE, Larsen MH, Robbe-austerman S, Williams MC, Palmer MV. Emerging tuberculosis pathogen hijacks social communication behavior in the group-living banded mongoose (Mungos mungo). mBio. 2016;7:3. doi:10.1128/mBio.00281-16
  • Henderson RA, Watkins SC, Flynn JL. Activation of human dendritic cells following infection with Mycobacterium tuberculosis. J Immunol. 1997;159(2):635–643.9218578
  • Ganbat D, Seehase S, Richter E, et al. Mycobacteria infect different cell types in the human lung and cause species dependent cellular changes in infected cells. BMC Pulm Med. 2016:16. doi:10.1186/s12890-016-0185-5.26801409
  • Divangahi M, Mostowy S, Coulombe F, et al. NOD2-deficient mice have impaired resistance to mycobacterium tuberculosis infection through defective innate and adaptive immunity. J Immunol. 2008;181(10):7157–7165. doi:10.4049/jimmunol.181.10.715718981137
  • Flannagan RS, Cosío G, Grinstein S. Antimicrobial mechanisms of phagocytes and bacterial evasion strategies. Nat Rev Microbiol. 2009;7(5):355–366. doi:10.1038/nrmicro212819369951
  • Philips JA, Ernst JD. Tuberculosis pathogenesis and Immunity. Annu Rev Pathol Mech Dis. 2012;7(1):353–384. doi:10.1146/annurev-pathol-011811-132458
  • Schnettger L, Rodgers A, Repnik U, et al. A Rab20-dependent membrane trafficking pathway controls M. tuberculosis replication by regulating phagosome spaciousness and integrity. Cell Host Microbe. 2017;21(5):619–628.e5. doi:10.1021/es103418828494243
  • Tăbăran A-F, Cornel C. Macrophages targeted drug delivery as a key therapy in infectious disease. Biotechnol Mol Biol Nanomedicine. 2014;2:1.
  • Sturgill-Koszycki S, Schlesinger PH, Chakraborty P, et al. Lack of acidification in Mycobacterium phagosomes produced by exclusion of the vesicular proton-ATPase. Science. 1994;263(5147):678–681. doi:10.1126/science.83032778303277
  • Stanley LR, Vinay K, Abul K, Ramzi S, Nelson F. Robbins & Cotran Pathologic Basis of Disease. Saunders/Elsevier; 2010.
  • Ferrari G, Langen H, Naito M, Pieters J. A coat protein on phagosomes involved in the intracellular survival of mycobacteria. Cell. 1999;97(4):435–447. doi:10.1016/S0092-8674(00)80754-010338208
  • Hmama Z, Sendide K, Talal A, Garcia R, Dobos K, Reiner NE. Quantitative analysis of phagolysosome fusion in intact cells: inhibition by mycobacterial lipoarabinomannan and rescue by an 1α, 25-dihydroxyvitamin D3–phosphoinositide 3-kinase pathway. J Cell Sci. 2004;117(10):2131–2140. doi:10.1242/jcs.0107215090599
  • Pabst M, Gross J, Brozna J, Goren M. Inhibition of macrophage priming by sulfatide from Mycobacterium tuberculosis. J Immunol. 1988;140(2):634–640.2826597
  • Vergne I, Chua J, Deretic V. Tuberculosis toxin blocking phagosome maturation inhibits a novel Ca2+/calmodulin-PI3K hVPS34 cascade. J Exp Med. 2003;198(4):653–659. doi:10.1084/jem.2003052712925680
  • Clemens DL, Horwitz MA. The Mycobacterium tuberculosis phagosome interacts with early endosomes and is accessible to exogenously administered transferrin. J Exp Med. 1996;184(4):1349–1355. doi:10.1084/jem.184.4.13498879207
  • Lyadova IV, Panteleev AV. Th1 and Th17 cells in tuberculosis: protection, pathology, and biomarkers. Mediators Inflamm. 2015;2015:1–13. doi:10.1155/2015/854507
  • Ashenafi S, Aderaye G, Bekele A, et al. Progression of clinical tuberculosis is associated with a Th2 immune response signature in combination with elevated levels of SOCS3. Clin Immunol. 2014;151(2):84–99. doi:10.1016/j.clim.2014.01.01024584041
  • Infante-Duarto C, Kamradt T Thl/Th2 balance in infection.:22.
  • Mohareer K, Asalla S, Banerjee S. Cell death at the cross roads of host-pathogen interaction in Mycobacterium tuberculosis infection. Tuberculosis. 2018;113:99–121. doi:10.1016/j.tube.2018.09.00730514519
  • Lerner TR, Borel S, Greenwood DJ, et al. Mycobacterium tuberculosis replicates within necrotic human macrophages. J Cell Biol. 2017;216(3):583–594. doi:10.1161/CIRCRESAHA.117.31140128242744
  • Dallenga T, Repnik U, Corleis B, et al. M. tuberculosis-induced necrosis of infected neutrophils promotes bacterial growth following phagocytosis by macrophages. Cell Host Microbe. 2017;22(4):519–530.e3. doi:10.1016/j.chom.2017.09.00329024644
  • Queval CJ, Brosch R, Simeone R. The macrophage: a disputed fortress in the battle against mycobacterium tuberculosis. Front Microbiol. 2017;8. doi:10.3389/fmicb.2017.0228428144237
  • Shanmugasundaram T, Radhakrishnan M, Gopikrishnan V, Pazhanimurugan R, Balagurunathan R. A study of the bactericidal, anti-biofouling, cytotoxic and antioxidant properties of actinobacterially synthesised silver nanoparticles. Colloids Surf B Biointerfaces. 2013;111:680–687. doi:10.1016/j.colsurfb.2013.06.04523911625
  • Aderibigbe BA. Metal-based nanoparticles for the treatment of infectious diseases. Mol Basel Switz. 2017;22:8. doi:10.3390/molecules22081370
  • Fathima JB, Pugazhendhi A, Venis R. Synthesis and characterization of ZrO2 nanoparticles-antimicrobial activity and their prospective role in dental care. Microb Pathog. 2017;110:245–251. doi:10.1016/j.micpath.2017.06.03928666841
  • Shanmuganathan R, LewisOscar F, Shanmugam S, et al. Core/shell nanoparticles: synthesis, investigation of antimicrobial potential and photocatalytic degradation of Rhodamine B. J Photochem Photobiol B. 2020;202:111729. doi:10.1016/j.jphotobiol.2019.11172931835161
  • Pugazhendhi A, Kumar SS, Manikandan M, Saravanan M. Photocatalytic properties and antimicrobial efficacy of Fe doped CuO nanoparticles against the pathogenic bacteria and fungi. Microb Pathog. 2018;122:84–89. doi:10.1016/j.micpath.2018.06.01629894807
  • Vasantharaj S, Sathiyavimal S, Senthilkumar P, LewisOscar F, Pugazhendhi A. Biosynthesis of iron oxide nanoparticles using leaf extract of Ruellia tuberosa: antimicrobial properties and their applications in photocatalytic degradation. J Photochem Photobiol B. 2019;192:74–82. doi:10.1016/j.jphotobiol.2018.12.02530685586
  • Reznickova A, Slavikova N, Kolska Z, et al. PEGylated gold nanoparticles: stability, cytotoxicity and antibacterial activity. Colloids Surf Physicochem Eng Asp. 2019;560:26–34. doi:10.1016/j.colsurfa.2018.09.083
  • Mocan L, Matea C, Tabaran FA, et al. Selective in vitro photothermal nano-therapy of MRSA infections mediated by IgG conjugated gold nanoparticles. Sci Rep. 2016:6. doi:10.1038/srep39466.28442741
  • Jeyarani S, Vinita NM, Puja P, et al. Biomimetic gold nanoparticles for its cytotoxicity and biocompatibility evidenced by fluorescence-based assays in cancer (MDA-MB-231) and non-cancerous (HEK-293) cells. J Photochem Photobiol B. 2020;202:111715. doi:10.1016/j.jphotobiol.2019.11171531790882
  • Chellapandian C, Ramkumar B, Puja P, Shanmuganathan R, Pugazhendhi A, Kumar P. Gold nanoparticles using red seaweed Gracilaria verrucosa: green synthesis, characterization and biocompatibility studies. Process Biochem. 2019;80:58–63. doi:10.1016/j.procbio.2019.02.009
  • Pugazhendhi A, Prabhu R, Muruganantham K, Shanmuganathan R, Natarajan S. Anticancer, antimicrobial and photocatalytic activities of green synthesized magnesium oxide nanoparticles (MgONPs) using aqueous extract of Sargassum wightii. J Photochem Photobiol B. 2019;190:86–97. doi:10.1016/j.jphotobiol.2018.11.01430504053
  • Hariharan D, Thangamuniyandi P, Jegatha Christy A, et al. Enhanced photocatalysis and anticancer activity of green hydrothermal synthesized Ag@TiO2 nanoparticles. J Photochem Photobiol B. 2020;202:111636. doi:10.1016/j.jphotobiol.2019.11163631739259
  • Hariharan D, Thangamuniyandi P, Selvakumar P, et al. Green approach synthesis of Pd@TiO2 nanoparticles: characterization, visible light active picric acid degradation and anticancer activity. Process Biochem. 2019;87:83–88. doi:10.1016/j.procbio.2019.09.024
  • Ramkumar VS, Pugazhendhi A, Gopalakrishnan K, et al. Biofabrication and characterization of silver nanoparticles using aqueous extract of seaweed Enteromorpha compressa and its biomedical properties. Biotechnol Rep. 2017;14:1–7. doi:10.1016/j.btre.2017.02.001
  • Madhubala V, Pugazhendhi A, Thirunavukarasu K. Cytotoxic and immunomodulatory effects of the low concentration of titanium dioxide nanoparticles (TiO2 NPs) on human cell lines - An in vitro study. Process Biochem. 2019;86:186–195. doi:10.1016/j.procbio.2019.08.004
  • Fathima JB, Pugazhendhi A, Oves M, Venis R. Synthesis of eco-friendly copper nanoparticles for augmentation of catalytic degradation of organic dyes. J Mol Liq. 2018;260:1–8. doi:10.1016/j.molliq.2018.03.033
  • Vasantharaj S, Sathiyavimal S, Saravanan M, et al. Synthesis of ecofriendly copper oxide nanoparticles for fabrication over textile fabrics: characterization of antibacterial activity and dye degradation potential. J Photochem Photobiol B. 2019;191:143–149. doi:10.1016/j.jphotobiol.2018.12.02630639996
  • Saratale RG, Ghodake GS, Shinde SK, et al. Photocatalytic activity of CuO/Cu(OH)2 nanostructures in the degradation of Reactive Green 19A and textile effluent, phytotoxicity studies and their biogenic properties (antibacterial and anticancer). J Environ Manage. 2018;223:1086–1097. doi:10.1016/j.jenvman.2018.04.07229735295
  • Varadavenkatesan T, Lyubchik E, Pai S, Pugazhendhi A, Vinayagam R, Selvaraj R. Photocatalytic degradation of Rhodamine B by zinc oxide nanoparticles synthesized using the leaf extract of Cyanometra ramiflora. J Photochem Photobiol B. 2019;199:111621. doi:10.1016/j.jphotobiol.2019.11162131610434
  • Shanmuganathan R, Edison TNJI, LewisOscar F, Kumar P, Shanmugam S, Pugazhendhi A. Chitosan nanopolymers: an overview of drug delivery against cancer. Int J Biol Macromol. 2019;130:727–736. doi:10.1016/j.ijbiomac.2019.02.06030771392
  • Sanpui P, Chattopadhyay A, Ghosh SS. Induction of apoptosis in cancer cells at low silver nanoparticle concentrations using chitosan nanocarrier. ACS Appl Mater Interfaces. 2011;3(2):218–228. doi:10.1021/am100840c21280584
  • Suganya M, Gnanamangai BM, Govindasamy C, et al. Mitochondrial dysfunction mediated apoptosis of HT-29 cells through CS-PAC-AgNPs and investigation of genotoxic effects in zebra (Danio rerio) fish model for drug delivery. Saudi J Biol Sci. 2019;26(4):767–776. doi:10.1016/j.sjbs.2019.03.00731049002
  • Russell AD, Hugo WB. 7 Antimicrobial activity and action of silver In: Progress in Medicinal Chemistry. Vol. 31 Elsevier;1994::351–370. doi:10.1016/S0079-6468(08)70024-9
  • Silva JP, Appelberg R, Gama FM. Antimicrobial peptides as novel anti-tuberculosis therapeutics. Biotechnol Adv. 2016;34(5):924–940. doi:10.1016/j.biotechadv.2016.05.00727235189
  • Khan F, Khan MM, Kim Y-M. Recent progress and future perspectives of antibiofilm drugs immobilized on nanomaterials. Curr Pharm Biotechnol. 2018;19(8):631–643. doi:10.2174/138920101966618082809005230152281
  • Raffi M, Mehrwan S, Bhatti TM, et al. Investigations into the antibacterial behavior of copper nanoparticles against Escherichia coli. Ann Microbiol. 2010;60(1):75–80. doi:10.1007/s13213-010-0015-6
  • Pham DTN, Khan F, Phan TTV, et al. Biofilm inhibition, modulation of virulence and motility properties by FeOOH nanoparticle in Pseudomonas aeruginosa. Braz J Microbiol. 2019;50(3):791–805. doi:10.1007/s42770-019-00108-z31250405
  • Khan F, Manivasagan P, Lee J-W, Pham DTN, Oh J, Kim Y-M. Fucoidan-stabilized gold nanoparticle-mediated biofilm inhibition, attenuation of virulence and motility properties in Pseudomonas aeruginosa PAO1. Mar Drugs. 2019;17:4. doi:10.3390/md17040208
  • Javaid A, Oloketuyi SF, Khan MM, Khan F. Diversity of bacterial synthesis of silver nanoparticles. BioNanoScience. 2018;8(1):43–59. doi:10.1007/s12668-017-0496-x
  • Mocan T, Matea CT, Pop T, et al. Carbon nanotubes as anti-bacterial agents. Cell Mol Life Sci CMLS. 2017;74(19):3467–3479. doi:10.1007/s00018-017-2532-y28536787
  • Khan F, Pham DTN, Oloketuyi SF, Manivasagan P, Oh J, Kim Y-M. Chitosan and their derivatives: antibiofilm drugs against pathogenic bacteria. Colloids Surf B Biointerfaces. 2020;185:110627. doi:10.1016/j.colsurfb.2019.11062731732391
  • Khan F, Manivasagan P, Pham DTN, Oh J, Kim S-K, Kim Y-M. Antibiofilm and antivirulence properties of chitosan-polypyrrole nanocomposites to Pseudomonas aeruginosa. Microb Pathog. 2019;128:363–373. doi:10.1016/j.micpath.2019.01.03330684638
  • Manivasagan P, Khan F, Hoang G, et al. Thiol chitosan-wrapped gold nanoshells for near-infrared laser-induced photothermal destruction of antibiotic-resistant bacteria. Carbohydr Polym. 2019;225:115228. doi:10.1016/j.carbpol.2019.11522831521288
  • Zazo H, Colino CI, Lanao JM. Current applications of nanoparticles in infectious diseases. J Control Release off J Control Release Soc. 2016;224:86–102. doi:10.1016/j.jconrel.2016.01.008
  • Costa-gouveia J, Aínsa JA, Brodin P, Lucía A. How can nanoparticles contribute to antituberculosis therapy? Drug Discov Today. 2017;22(3):600–607. doi:10.1016/j.drudis.2017.01.01128137645
  • Kreytsberg GN, Gracheva IE, Kibrik BS, Golikov IV. Antituberculous effect of silver nanoparticles. J Phys Conf Ser. 2011;291:012030. doi:10.1088/1742-6596/291/1/012030
  • Song B, Zhang C, Zeng G, Gong J, Chang Y, Jiang Y. Antibacterial properties and mechanism of graphene oxide-silver nanocomposites as bactericidal agents for water disinfection. Arch Biochem Biophys. 2016;604:167–176. doi:10.1016/j.abb.2016.04.01827170600
  • Liu C, Guo J, Yan X, et al. Antimicrobial nanomaterials against biofilms: an alternative strategy. Environ Rev. 2016;25(2):225–244. doi:10.1139/er-2016-0046
  • Franci G, Falanga A, Galdiero S, et al. Silver nanoparticles as potential antibacterial agents. Mol Basel Switz. 2015;20(5):8856–8874. doi:10.3390/molecules20058856
  • Dakal TC, Kumar A, Majumdar RS, Yadav V. Mechanistic basis of antimicrobial actions of silver nanoparticles. Front Microbiol. 2016;7:1831. doi:10.3389/fmicb.2016.0183127899918
  • Morones JR, Elechiguerra JL, Camacho A, et al. The bactericidal effect of silver nanoparticles. Nanotechnology. 2005;16(10):2346. doi:10.1088/0957-4484/16/10/05920818017
  • Kumar DA, Palanichamy V, Roopan SM. Green synthesis of silver nanoparticles using Alternanthera dentata leaf extract at room temperature and their antimicrobial activity. Spectrochim Acta A Mol Biomol Spectrosc. 2014;127:168–171. doi:10.1016/j.saa.2014.02.05824632169
  • Jain J, Arora S, Rajwade JM, Omray P, Khandelwal S, Paknikar KM. Silver nanoparticles in therapeutics: development of an antimicrobial gel formulation for topical use. Mol Pharm. 2009;6(5):1388–1401. doi:10.1021/mp900056g19473014
  • Le Ouay B, Stellacci F. Antibacterial activity of silver nanoparticles: a surface science insight. Nano Today. 2015;10(3):339–354. doi:10.1016/j.nantod.2015.04.002
  • Huh AJ, Kwon YJ. “Nanoantibiotics”: a new paradigm for treating infectious diseases using nanomaterials in the antibiotics resistant era. J Control Release off J Control Release Soc. 2011;156(2):128–145. doi:10.1016/j.jconrel.2011.07.002
  • Kim JS, Kuk E, Yu KN, et al. Antimicrobial effects of silver nanoparticles. Nanomedicine Nanotechnol Biol Med. 2007;3(1):95–101. doi:10.1016/j.nano.2006.12.001
  • Al-Sharqi A, Apun K, Vincent M, Kanakaraju D, Bilung LM. Enhancement of the antibacterial efficiency of silver nanoparticles against gram-positive and gram-negative bacteria using blue laser light. Int J Photoenergy. doi:10.1155/2019/2528490
  • Velayati AA, Farnia P, Ibrahim TA, et al. Differences in cell wall thickness between resistant and nonresistant strains of Mycobacterium tuberculosis: using transmission electron microscopy. Chemotherapy. 2009;55(5):303–307. doi:10.1159/00022642519556787
  • Mai-Prochnow A, Clauson M, Hong J, Murphy AB. Gram positive and Gram negative bacteria differ in their sensitivity to cold plasma. Sci Rep. 2016;6. doi:10.1038/srep3861028442741
  • Hett EC, Rubin EJ. Bacterial growth and cell division: a mycobacterial perspective. Microbiol Mol Biol Rev MMBR. 2008;72(1):126–156. doi:10.1128/MMBR.00028-0718322037
  • Saravanan M, Barik SK, MubarakAli D, Prakash P, Pugazhendhi A. Synthesis of silver nanoparticles from Bacillus brevis (NCIM 2533) and their antibacterial activity against pathogenic bacteria. Microb Pathog. 2018;116:221–226. doi:10.1016/j.micpath.2018.01.03829407231
  • Seth D, Sarkar A, Mitra D. Nanomedicine to counter syndemic tuberculosis and HIV infection: current knowledge and state of art. Nanosci Nanoeng. 2014;9.
  • Abdel-Aziz MM, Elella MHA, Mohamed RR. Green synthesis of quaternized chitosan/silver nanocomposites for targeting mycobacterium tuberculosis and lung carcinoma cells (A-549). Int J Biol Macromol. 2019. doi:10/ggdd6n
  • El Badawy AM, Silva RG, Morris B, Scheckel KG, Suidan MT, Tolaymat TM. Surface charge-dependent toxicity of silver nanoparticles. Environ Sci Technol. 2011;45(1):283–287. doi:10/cc29h521133412
  • Sondi I, Salopek-Sondi B. Silver nanoparticles as antimicrobial agent: a case study on E. coli as a model for Gram-negative bacteria. J Colloid Interface Sci. 2004;275(1):177–182. doi:10.1016/j.jcis.2004.02.01215158396
  • Cho K-H, Park J-E, Osaka T, Park S-G. The study of antimicrobial activity and preservative effects of nanosilver ingredient. Electrochim Acta. 2005;51(5):956–960. doi:10.1016/j.electacta.2005.04.071
  • McQuillan JS, Groenaga Infante H, Stokes E, Shaw AM. Silver nanoparticle enhanced silver ion stress response in Escherichia coli K12. Nanotoxicology. 2012;6(8):857–866. doi:10.3109/17435390.2011.62653222007647
  • Orlov IA, Sankova TP, Babich PS, et al. New silver nanoparticles induce apoptosis-like process in E. coli and interfere with mammalian copper metabolism. Int J Nanomedicine. doi:10.2147/IJN.S117745
  • Rodgers FG, Tzianabos AO, Elliott TSJ. The effect of antibiotics that inhibit cell-wall, protein, and DNA synthesis on the growth and morphology of Legionella pneumophila. J Med Microbiol. 1990;31(1):37–44. doi:10.1099/00222615-31-1-372296040
  • van der Wal A, Norde W, Zehnder AJB, Lyklema J. Determination of the total charge in the cell walls of Gram-positive bacteria. Colloids Surf B Biointerfaces. 1997;9(1–2):81–100. doi:10.1016/S0927-7765(96)01340-9
  • Shaik M, Albalawi G, Khan S, et al. “Miswak” based green synthesis of silver nanoparticles: evaluation and comparison of their microbicidal activities with the chemical synthesis. Molecules. 2016;21(11):1478. doi:10.3390/molecules21111478
  • Ashraf S, Akhtar N, Ghauri M, Rajoka M, Khalid ZM, Hussain I. Polyhexamethylene biguanide functionalized cationic silver nanoparticles for enhanced antimicrobial activity. Nanoscale Res Lett. 2012;7(1):267. doi:10.1186/1556-276X-7-26722625664
  • Ivask A, ElBadawy A, Kaweeteerawat C, et al. Toxicity mechanisms in escherichia coli vary for silver nanoparticles and differ from ionic silver. ACS Nano. 2014;8(1):374–386. doi:10/f5qqhz24341736
  • Kvítek L, Panáček A, Soukupová J, et al. Effect of surfactants and polymers on stability and antibacterial activity of silver nanoparticles (NPs). J Phys Chem C. 2008;112(15):5825–5834. doi:10.1021/jp711616v
  • Yin R, Agrawal T, Khan U, et al. Antimicrobial photodynamic inactivation in nanomedicine: small light strides against bad bugs. Nanomed. 2015;10(15):2379–2404. doi:10.2217/nnm.15.67
  • Yun’an Qing LC, Li R, Liu G, et al. Potential antibacterial mechanism of silver nanoparticles and the optimization of orthopedic implants by advanced modification technologies. Int J Nanomedicine. 2018;13:3311. doi:10.2147/IJN.S16512529892194
  • Bonnet M, Massard C, Veisseire P, Camares O, Awitor KO. Environmental toxicity and antimicrobial efficiency of titanium dioxide nanoparticles in suspension. J Biomater Nanobiotech. 2015;6(3):213–224. doi:10.4236/jbnb.2015.63020
  • Parikh AN, Gillmor SD, Beers JD, Beardmore KM, Cutts RW, Swanson BI. Characterization of chain molecular assemblies in long-chain, layered silver thiolates: a joint infrared spectroscopy and X-ray diffraction study. J Phys Chem B. 1999;103(15):2850–2861. doi:10.1021/jp983938b
  • Dobias J, Bernier-Latmani R. Silver release from silver nanoparticles in natural waters. Environ Sci Technol. 2013;47(9):4140–4146. doi:10.1021/es304023p23517230
  • Park E-J, Yi J, Kim Y, Choi K, Park K. Silver nanoparticles induce cytotoxicity by a Trojan-horse type mechanism. Toxicol in Vitro. 2010;24(3):872–878. doi:10.1016/j.tiv.2009.12.00119969064
  • He D, Miller CJ, Waite TD. Fenton-like zero-valent silver nanoparticle-mediated hydroxyl radical production. J Catal. 2014;317:198–205. doi:10.1016/j.jcat.2014.06.016
  • Sisubalan N, Ramkumar VS, Pugazhendhi A, et al. ROS-mediated cytotoxic activity of ZnO and CeO2 nanoparticles synthesized using the Rubia cordifolia L. leaf extract on MG-63 human osteosarcoma cell lines. Environ Sci Pollut Res. 2018;25(11):10482–10492. doi:10.1007/s11356-017-0003-5
  • Knaapen AM, Borm PJA, Albrecht C, Schins RPF. Inhaled particles and lung cancer. Part A: mechanisms. Int J Cancer. 2004;109(6):799–809. doi:10.1002/ijc.1170815027112
  • Samuel MS, Jose S, Selvarajan E, Mathimani T, Pugazhendhi A. Biosynthesized silver nanoparticles using Bacillus amyloliquefaciens; application for cytotoxicity effect on A549 cell line and photocatalytic degradation of p-nitrophenol. J Photochem Photobiol B. 2020;202:111642. doi:10.1016/j.jphotobiol.2019.11164231734434
  • Speight JG, editor. Redox Transformations In: Reaction Mechanisms in Environmental Engineering. Elsevier; 2018:231–267. doi:10.1016/B978-0-12-804422-3.00007-9
  • Forrester SJ, Kikuchi DS, Hernandes MS, Xu Q, Griendling KK. Reactive oxygen species in metabolic and inflammatory signaling. Circ Res. 2018;122(6):877–902. doi:10.1161/CIRCRESAHA.117.31140129700084
  • Zielinska E, Tukaj C, Radomski MW, Inkielewicz-stepniak I. Molecular mechanism of silver nanoparticles-induced human osteoblast cell death: protective effect of inducible nitric oxide synthase inhibitor. PLoS One. 2016;11(10):e0164137. doi:10/f9rvdx27716791
  • Bressan E, Ferroni L, Gardin C, et al. Silver nanoparticles and mitochondrial interaction. Int J Dent. 2013;2013:1–8. doi:10.1155/2013/312747
  • Maurer LL, Meyer JN. A systematic review of evidence for silver nanoparticle-induced mitochondrial toxicity. Environ Sci Nano. 2016;3(2):311–322. doi:10.1039/C5EN00187K
  • Kim S, Choi JE, Choi J, et al. Oxidative stress-dependent toxicity of silver nanoparticles in human hepatoma cells. Toxicol in Vitro. 2009;23(6):1076–1084. doi:10.1016/j.tiv.2009.06.00119508889
  • Miyayama T, Matsuoka M. Involvement of lysosomal dysfunction in silver nanoparticle-induced cellular damage in A549 human lung alveolar epithelial cells. J Occup Med Toxicol. 2016;11(1):1. doi:10.1186/s12995-016-0090-026759602
  • Park MVDZ, Neigh AM, Vermeulen JP, et al. The effect of particle size on the cytotoxicity, inflammation, developmental toxicity and genotoxicity of silver nanoparticles. Biomaterials. 2011;32(36):9810–9817. doi:10.1016/j.biomaterials.2011.08.08521944826
  • Singh R, Nawale L, Arkile M, et al. Phytogenic silver, gold, and bimetallic nanoparticles as novel antitubercular agents. Int J Nanomedicine. 2016;11:1889.27217751
  • Xiu Z, Zhang Q, Puppala HL, Colvin VL, Alvarez PJJ. Negligible particle-specific antibacterial activity of silver nanoparticles. Nano Lett. 2012;12(8):4271–4275. doi:10.1021/nl301934w22765771
  • Behra R, Sigg L, Clift MJD, et al. Bioavailability of silver nanoparticles and ions: from a chemical and biochemical perspective. J R Soc Interface. 2013;10:87. doi:10.1098/rsif.2013.0396
  • Saratale RG, Karuppusamy I, Saratale GD, et al. A comprehensive review on green nanomaterials using biological systems: recent perception and their future applications. Colloids Surf B Biointerfaces. 2018;170:20–35. doi:10.1016/j.colsurfb.2018.05.04529860217
  • Saratale RG, Saratale GD, Shin HS, et al. New insights on the green synthesis of metallic nanoparticles using plant and waste biomaterials: current knowledge, their agricultural and environmental applications. Environ Sci Pollut Res. 2018;25(11):10164–10183. doi:10.1007/s11356-017-9912-6
  • Kote JR, Kadam AS, Patil SS, Mane RS. Green functionalized silver nanoparticles with significantly enhanced antimycobactericidal and cytotoxicity performances of asparagus racemosus Linn. Int J New Technol Sci. 2016;3(2):15.
  • Roy A, Bulut O, Some S, Mandal AK, Yilmaz MD. Green synthesis of silver nanoparticles: biomolecule-nanoparticle organizations targeting antimicrobial activity. RSC Adv. 2019;9(5):2673–2702. doi:10.1039/C8RA08982E
  • Kerry RG, Gouda S, Sil B, et al. Cure of tuberculosis using nanotechnology: an overview. J Microbiol. 2018;56(5):287–299. doi:10.1007/s12275-018-7414-y29721825
  • Arokiyaraj S, Arasu MV, Vincent S, et al. Rapid green synthesis of silver nanoparticles from Chrysanthemum indicum L and its antibacterial and cytotoxic effects: an in vitro study. Int J Nanomedicine. doi:10.2147/IJN.S53546
  • Ahmed S, Ahmad M, Swami BL, Ikram S. A review on plants extract mediated synthesis of silver nanoparticles for antimicrobial applications: a green expertise. J Adv Res. 2016;7(1):17–28. doi:10.1016/j.jare.2015.02.00726843966
  • Shaik M, Khan M, Kuniyil M, et al. Plant-extract-assisted green synthesis of silver nanoparticles using origanum vulgare L. Extract and their microbicidal activities. Sustainability. 2018;10(4):913. doi:10.3390/su10040913
  • Pugazhendhi A, Prabakar D, Jacob JM, Karuppusamy I, Saratale RG. Synthesis and characterization of silver nanoparticles using Gelidium amansii and its antimicrobial property against various pathogenic bacteria. Microb Pathog. 2018;114:41–45. doi:10.1016/j.micpath.2017.11.01329146498
  • Oves M, Aslam M, Rauf MA, et al. Antimicrobial and anticancer activities of silver nanoparticles synthesized from the root hair extract of Phoenix dactylifera. Mater Sci Eng C. 2018;89:429–443. doi:10.1016/j.msec.2018.03.035
  • Singh R, Nawale LU, Arkile M, et al. Chemical and biological metal nanoparticles as antimycobacterial agents: a comparative study. Int J Antimicrob Agents. 2015;46(2):183–188. doi:10.1016/j.ijantimicag.2015.03.01426009020
  • Rónavári A, Kovács D, Igaz N, et al. Biological activity of green-synthesized silver nanoparticles depends on the applied natural extracts: a comprehensive study. Int J Nanomedicine. 2017;12:871–883. doi:10.2147/IJN.S12284228184158
  • Rai M, Ingle AP, Pandit R, et al. Broadening the spectrum of small-molecule antibacterials by metallic nanoparticles to overcome microbial resistance. Int J Pharm. 2017;532(1):139–148. doi:10.1016/j.ijpharm.2017.08.12728870767
  • Baptista PV, McCusker MP, Carvalho A, et al. Nano-strategies to fight multidrug resistant bacteria—“a battle of the titans. Front Microbiol. 2018;9:1441. doi:10.3389/fmicb.2018.0144130013539
  • Djafari J, Marinho C, Santos T, et al. New synthesis of gold- and silver-based nano-tetracycline composites. ChemistryOpen. 2016;5(3):206–212. doi:10.1002/open.20160001627957408
  • Deng H, McShan D, Zhang Y, et al. Mechanistic study of the synergistic antibacterial activity of combined silver nanoparticles and common antibiotics. Environ Sci Technol. 2016;50(16):8840–8848. doi:10.1021/acs.est.6b0099827390928
  • Birla SS, Tiwari VV, Gade AK, Ingle AP, Yadav AP, Rai MK. Fabrication of silver nanoparticles by Phoma glomerata and its combined effect against Escherichia coli, Pseudomonas aeruginosa and Staphylococcus aureus. Lett Appl Microbiol. 2009;48(2):173–179. doi:10.1111/j.1472-765X.2008.02510.x19141039
  • Brown AN, Smith K, Samuels TA, Lu J, Obare SO, Scott ME. Nanoparticles functionalized with ampicillin destroy multiple-antibiotic-resistant isolates of pseudomonas aeruginosa and enterobacter aerogenes and methicillin-resistant Staphylococcus aureus. Appl Environ Microbiol. 2012;78(8):2768–2774. doi:10.1128/AEM.06513-1122286985
  • Saratale GD, Saratale RG, Benelli G, et al. Anti-diabetic potential of silver nanoparticles synthesized with argyreia nervosa leaf extract high synergistic antibacterial activity with standard antibiotics against foodborne bacteria. J Clust Sci. 2017;28(3):1709–1727. doi:10.1007/s10876-017-1179-z
  • Shanmuganathan R, MubarakAli D, Prabakar D, et al. An enhancement of antimicrobial efficacy of biogenic and ceftriaxone-conjugated silver nanoparticles: green approach. Environ Sci Pollut Res. 2018;25(11):10362–10370. doi:10.1007/s11356-017-9367-9
  • Hwang I-S, Hwang JH, Choi H, Kim K-J, Lee DG. Synergistic effects between silver nanoparticles and antibiotics and the mechanisms involved. J Med Microbiol. 2012;61(Pt_12):1719–1726. doi:10.1099/jmm.0.047100-022956753
  • Farooq U, Ahmad T, Khan A, et al. Rifampicin conjugated silver nanoparticles: a new arena for development of antibiofilm potential against methicillin resistant Staphylococcus aureus and Klebsiella pneumoniae. Int J Nanomedicine. 2019;14:3983–3993. doi:10.2147/IJN.S19819431213810
  • Li P, Li J, Wu C, Wu Q, Li J. Synergistic antibacterial effects of β-lactam antibiotic combined with silver nanoparticles. Nanotechnology. 2005;16(9):1912–1917. doi:10.1088/0957-4484/16/9/082
  • Arakha M, Jha S. Effects of photocatalytic nanoparticle interfaces on biological membranes and biomacromolecules. 2017. doi:10/ggdp7k
  • Nasiruddin M, Neyaz M, Das S. Nanotechnology-based approach in tuberculosis treatment. Tuberc Res Treat. 2017;2017. doi:10.1155/2017/4920209.
  • Singh R, Nawale L, Arkile M, et al. Phytogenic silver, gold, and bimetallic nanoparticles as novel antitubercular agents. Int J Nanomedicine. 2016;11:1889–1897. doi:10.2147/IJN.S10248827217751
  • Song HY, Ko KK, Oh IH, Lee BT. Fabrication of silver nanoparticles and their antimicrobial mechanisms. Eur Cell Mater. 2006;11(suppl 1):58.
  • Jaryal N, Kaur H. Plumbago auriculata leaf extract-mediated AgNPs and its activities as antioxidant, anti-TB and dye degrading agents. J Biomater Sci Polym Ed. 2017;28(16):1847–1858. doi:10.1080/09205063.2017.135467328697688
  • Paarakh PM. Anti-tubercular activity of silver nanoparticle synthesized from the fruits of coriandrum sativum linn. World J Pharm Pharm Sci. 2017;1720–1727. doi:10/ggdkzz
  • Raja A, Salique SM, Gajalakshmi P, James A. Antibacterial and hemolytic activity of green silver nanoparticles from Catharanthus roseus. Int J Pharm Sci Nanotechnol. 2016;9(1):7.
  • Kote JR, Mulani RM, Kadam AS, Solankar BM. Anti-Mycobacterial Activity of Nanoparticles from Psidium Guajava L. 2014:5.
  • Daniel SCGK, Banu BN, Harshiny M, et al. Ipomea carnea -based silver nanoparticle synthesis for antibacterial activity against selected human pathogens. J Exp Nanosci. 2014;9(2):197–209. doi:10.1080/17458080.2011.654274
  • Banu A. Biosynthesis of monodispersed silver nanoparticles and their activity against Mycobacterium tuberculosis. J Nanomedicine Biotherapeutic Discov. 2013;03:01. doi:10.4172/2155-983X.1000110
  • Agarwal P, Mehta A, Kachhwaha S, Kothari SL. Green synthesis of silver nanoparticles and their activity against Mycobacterium tuberculosis. Adv Sci Eng Med. 2013;5(7):709–714. doi:10.1166/asem.2013.1307
  • Heidary M, Zaker Bostanabad S, Amini SM, et al. The anti-mycobacterial activity of Ag, ZnO, And Ag- ZnO nanoparticles against MDR- and XDR-Mycobacterium tuberculosis. Infect Drug Resist. 2019;12:3425–3435. doi:10.2147/IDR.S22140831807033
  • Ellis T, Chiappi M, García-trenco A, et al. Multimetallic microparticles increase the potency of rifampicin against intracellular Mycobacterium tuberculosis. ACS Nano. 2018;12(6):5228–5240. doi:10.1021/acsnano.7b0826429767993
  • Jafari A, Mosavari N, Movahedzadeh F, et al. Bactericidal impact of Ag, ZnO and mixed AgZnO colloidal nanoparticles on HRv Mycobacterium tuberculosis phagocytized by THP-1 cell lines. Microb Pathog. 2017;110:335–344. doi:10.1016/j.micpath.2017.07.01028710015
  • Jena P, Mohanty S, Mallick R, Jacob B, Sonawane A. Toxicity and antibacterial assessment of chitosancoated silver nanoparticles on human pathogens and macrophage cells. Int J Nanomedicine. 2012;7:1805. doi:10.2147/IJN.S3063122619529
  • Selim A, Elhaig MM, Taha SA, Nasr EA. Antibacterial activity of silver nanoparticles against field and reference strains of Mycobacterium tuberculosis, Mycobacterium bovis and multiple-drug-resistant tuberculosis strains. Rev Sci Tech OIE. 2018;37(3):823–830. doi:10.20506/rst.37.3.2888
  • Sivaraj A, Kumar V, Sunder R, Parthasarathy K, Kasivelu G. Commercial yeast extracts mediated green synthesis of silver chloride nanoparticles and their anti-mycobacterial activity. J Clust Sci. 2019. doi:10/ggdgtw
  • Kim J, Pitts B, Stewart PS, Camper A, Yoon J. Comparison of the antimicrobial effects of chlorine, silver ion, and tobramycin on biofilm. Antimicrob Agents Chemother. 2008;52(4):1446–1453. doi:10.1128/AAC.00054-0718195062
  • Patel S. Biogenic silver nanoparticles as potential agent against mycobacterium tuberculosis. Int J Res Appl Sci Eng Technol. 2018;6(1):505–511. doi:10.22214/ijraset.2018.1075
  • Seth D, Choudhury SR, Pradhan S, et al. Nature-inspired novel drug design paradigm using nanosilver: efficacy on multi-drug-resistant clinical isolates of tuberculosis. Curr Microbiol. 2011;62(3):715–726. doi:10.1007/s00284-010-9770-720936471
  • Punjabi K, Mehta S, Chavan R, Chitalia V, Deogharkar D, Deshpande S. Efficiency of biosynthesized silver and zinc nanoparticles against multi-drug resistant pathogens. Front Microbiol. 2018;9. doi:10/gfdvdk29387050
  • Sun F, Oh S, Kim J, et al. Enhanced internalization of macromolecular drugs into mycobacterium smegmatis with the assistance of silver nanoparticles. J Microbiol Biotechnol. 2017;27(8):1483–1490. doi:10.4014/jmb.1612.1204128595381
  • Padmaa MP. Green synthesis of silver nanoparticles using fruits of coriandrum sativum linn and its antioxidant activity. J Nat Prod Resour. 2015;1(1):19–22.
  • Donnellan S, Tran L, Johnston H, McLuckie J, Stevenson K, Stone V. A rapid screening assay for identifying mycobacteria targeted nanoparticle antibiotics. Nanotoxicology. 2016;10(6):761–769. doi:10.3109/17435390.2015.112446826618564
  • Jafari AR, Mosavi T, Mosavari N, et al. Mixed metal oxide nanoparticles inhibit growth of Mycobacterium tuberculosis into THP-1 cells. Int J Mycobacteriology. 2016;5:S181–S183. doi:10.1016/j.ijmyco.2016.09.011
  • Islam MS, Larimer C, Ojha A, Nettleship I. Antimycobacterial efficacy of silver nanoparticles as deposited on porous membrane filters. Mater Sci Eng C. 2013;33(8):4575–4581. doi:10.1016/j.msec.2013.07.013
  • Mohanty S, Mishra S, Jena P, Jacob B, Sarkar B, Sonawane A. An investigation on the antibacterial, cytotoxic, and antibiofilm efficacy of starch-stabilized silver nanoparticles. Nanomedicine Nanotechnol Biol Med. 2012;8(6):916–924. doi:10.1016/j.nano.2011.11.007
  • Zhou Y, Kong Y, Kundu S, Cirillo JD, Liang H. Antibacterial activities of gold and silver nanoparticles against Escherichia coli and bacillus Calmette-Guérin. J Nanobiotechnology. 2012;10(1):19. doi:10.1186/1477-3155-10-1922559747
  • Martinez-Gutierrez F, Olive PL, Banuelos A, et al. Synthesis, characterization, and evaluation of antimicrobial and cytotoxic effect of silver and titanium nanoparticles. Nanomedicine Nanotechnol Biol Med. 2010;6(5):681–688. doi:10.1016/j.nano.2010.02.001
  • Varghese MV, Dhumal RS, Patil SS, Paradkar AR, Khanna PK. Synthesis and in-vitro antimycobacterial studies of cysteine capped silver nano-particles. Synth React Inorg Met-Org Nano-Met Chem. 2009;39(9):554–558. doi:10.1080/15533170903327869
  • Uraskulova BB, Gyusan AO. The clinical and bacteriological study of the effectiveness of the application of silver nanoparticle for the treatment of tuberculosis. Vestn Otorinolaringol. 2017;82(3):54–57. doi:10.17116/otorino201782354-5728631683
  • Gmoshinski IV, Shumakova AA, Shipelin VA, Maltsev G, Khotimchenko SA. Influence of orally introduced silver nanoparticles on content of essential and toxic trace elements in organism. Nanotechnologies Russ. 2016;11(9–10):646–652. doi:10.1134/S1995078016050074
  • Zakharov AV, Khokhlov AL, Kibrik BS. Effectiveness of combination of isoniazid and silver nanoparticles in the treatment of experimental tuberculosis. Tuberc Lung Dis. 2017;95(6):51–58. doi:10.21292/2075-1230-2017-95-6-51-58
  • Zakharov AV, Khokhlov A. The results of experimental studies of the use of silver nanoparticles in tuberculosis drug-resistant pathogen. Med News North Cauc. 2019;14:1. doi:10/ggdjnk
  • Pugazhendhi A, Edison TNJI, Karuppusamy I, Kathirvel B. Inorganic nanoparticles: a potential cancer therapy for human welfare. Int J Pharm. 2018;539(1):104–111. doi:10.1016/j.ijpharm.2018.01.03429366941
  • Srinivasan M, Venkatesan M, Arumugam V, et al. Green synthesis and characterization of titanium dioxide nanoparticles (TiO2 NPs) using Sesbania grandiflora and evaluation of toxicity in zebrafish embryos. Process Biochem. 2019;80:197–202. doi:10.1016/j.procbio.2019.02.010
  • Vazquez-Muñoz R, Borrego B, Juárez-moreno K, et al. Toxicity of silver nanoparticles in biological systems: does the complexity of biological systems matter? Toxicol Lett. 2017;276:11–20. doi:10.1016/j.toxlet.2017.05.00728483428
  • Tarannum N, Divya K, Gautam Y. Facile green synthesis and applications of silver nanoparticles: a state-of-the-art review. RSC Adv. 2019;9(60):34926–34948. doi:10.1039/C9RA04164H
  • Dey Bhowmik A, Bandyopadhyay A, Chattopadhyay A. Cytotoxic and mutagenic effects of green silver nanoparticles in cancer and normal cells: a brief review. The Nucleus. 2019;62(3):277–285. doi:10.1007/s13237-019-00293-0
  • Saravanan M, Arokiyaraj S, Lakshmi T, Pugazhendhi A. Synthesis of silver nanoparticles from Phenerochaete chrysosporium (MTCC-787) and their antibacterial activity against human pathogenic bacteria. Microb Pathog. 2018;117:68–72. doi:10.1016/j.micpath.2018.02.00829427709
  • Yuan D, He H, Wu Y, Fan J, Cao Y. Physiologically based pharmacokinetic modeling of nanoparticles. J Pharm Sci. 2019;108(1):58–72. doi:10.1016/j.xphs.2018.10.03730385282
  • Ehlers S, Schaible UE. The granuloma in tuberculosis: dynamics of a host–pathogen collusion. Front Immunol. 2013;3. doi:10.3389/fimmu.2012.00411.
  • Sarkar S, Leo BF, Carranza C, et al. Modulation of human macrophage responses to mycobacterium tuberculosis by silver nanoparticles of different size and surface modification. PLoS One. 2015;10(11):e0143077. doi:10.1371/journal.pone.014307726580078
  • De Jong WH, Van Der Ven LTM, Sleijffers A, et al. Systemic and immunotoxicity of silver nanoparticles in an intravenous 28 days repeated dose toxicity study in rats. Biomaterials. 2013;34(33):8333–8343. doi:10.1016/j.biomaterials.2013.06.04823886731
  • Müller L, Steiner SK, Rodriguez-lorenzo L, Petri-fink A, Rothen-rutishauser B, Latzin P. Exposure to silver nanoparticles affects viability and function of natural killer cells, mostly via the release of ions. Cell Biol Toxicol. 2018;34(3):167–176. doi:10.1007/s10565-017-9403-z28721573
  • Alsaleh NB, Minarchick VC, Mendoza RP, Sharma B, Podila R, Brown JM. Silver nanoparticle immunomodulatory potential in absence of direct cytotoxicity in RAW 264.7 macrophages and MPRO 2.1 neutrophils. J Immunotoxicol. 2019;16(1):63–73. doi:10.1080/1547691X.2019.158892831282784
  • Yen H, Hsu S, Tsai C. Cytotoxicity and immunological response of gold and silver nanoparticles of different sizes. Small. 2009;5(13):1553–1561. doi:10.1002/smll.20090012619326357
  • Li T, Albee B, Alemayehu M, et al. Comparative toxicity study of Ag, Au, and Ag–Au bimetallic nanoparticles on Daphnia magna. Anal Bioanal Chem. 2010;398(2):689–700. doi:10.1007/s00216-010-3915-120577719
  • Suganthy N, Sri Ramkumar V, Pugazhendhi A, Benelli G, Archunan G. Biogenic synthesis of gold nanoparticles from Terminalia arjuna bark extract: assessment of safety aspects and neuroprotective potential via antioxidant, anticholinesterase, and antiamyloidogenic effects. Environ Sci Pollut Res. 2018;25(11):10418–10433. doi:10.1007/s11356-017-9789-4
  • Larimer C, Islam MS, Ojha A, Nettleship I. Mutation of environmental mycobacteria to resist silver nanoparticles also confers resistance to a common antibiotic. BioMetals. 2014;27(4):695–702. doi:10.1007/s10534-014-9761-424989695
  • Panáček A, Kvítek L, Smékalová M, et al. Bacterial resistance to silver nanoparticles and how to overcome it. Nat Nanotechnol. 2018;13(1):65–71. doi:10.1038/s41565-017-0013-y29203912
  • Muller M. Bacterial silver resistance gained by cooperative interspecies redox behavior. Antimicrob Agents Chemother. 2018;62:8. doi:10.1128/AAC.00672-18
  • Silver S. Bacterial silver resistance: molecular biology and uses and misuses of silver compounds. FEMS Microbiol Rev. 2003;27(2–3):341–353. doi:10.1016/S0168-6445(03)00047-012829274
  • Mijnendonckx K, Ali MM, Provoost A, et al. Spontaneous mutation in the AgrRS two-component regulatory system of Cupriavidus metallidurans results in enhanced silver resistance. Metallomics. 2019;11(11):1912–1924. doi:10.1039/C9MT00123A31609372
  • McDonnell G, Russell AD. Antiseptics and disinfectants: activity, action, and resistance. Clin Microbiol Rev. 1999;12(1):147–179. doi:10.1128/CMR.12.1.1479880479
  • Percival SL, Bowler PG, Russell D. Bacterial resistance to silver in wound care. J Hosp Infect. 2005;60(1):1–7. doi:10.1016/j.jhin.2004.11.01415823649
  • Woods EJ, Cochrane CA, Percival SL. Prevalence of silver resistance genes in bacteria isolated from human and horse wounds. Vet Microbiol. 2009;138(3–4):325–329. doi:10.1016/j.vetmic.2009.03.02319362435
  • Loh JV, Percival SL, Woods EJ, Williams NJ, Cochrane CA. Silver resistance in MRSA isolated from wound and nasal sources in humans and animals. Int Wound J. 2009;6(1):32–38. doi:10.1111/j.1742-481X.2008.00563.x19291113
  • Finley PJ, Norton R, Austin C, Mitchell A, Zank S, Durham P. Unprecedented silver resistance in clinically isolated enterobacteriaceae: major implications for burn and wound management. Antimicrob Agents Chemother. 2015;59(8):4734–4741. doi:10.1128/AAC.00026-1526014954
  • Percival SL, Woods E, Nutekpor M, Bowler P, Radford A, Cochrane C. Prevalence of silver resistance in bacteria isolated from diabetic foot ulcers and efficacy of silver-containing wound dressings. Ostomy Wound Manage. 2008;54(3):30–40.
  • Davis IJ, Richards H, Mullany P. Isolation of silver- and antibiotic-resistant Enterobacter cloacae from teeth. Oral Microbiol Immunol. 2005;20(3):191–194. doi:10.1111/j.1399-302X.2005.00218.x15836522
  • Summers AO, Wireman J, Vimy MJ, et al. Mercury released from dental “silver” fillings provokes an increase in mercury- and antibiotic-resistant bacteria in oral and intestinal floras of primates. Antimicrob Agents Chemother. 1993;37(4):825–834. doi:10.1128/AAC.37.4.8258280208
  • Haefeli C, Franklin C, Hardy K. Plasmid-determined silver resistance in Pseudomonas stutzeri isolated from a silver mine. J Bacteriol. 1984;158(1):389–392. doi:10.1128/JB.158.1.389-392.19846715284
  • Hanczvikkel A, Víg A, Tóth Á. Survival capability of healthcare-associated, multidrug-resistant bacteria on untreated and on antimicrobial textiles. J Ind Text. 2019;48(7):1113–1135. doi:10.1177/1528083718754901
  • Chapman JS. Disinfectant resistance mechanisms, cross-resistance, and co-resistance. Int Biodeterior Biodegrad. 2003;51(4):271–276. doi:10.1016/S0964-8305(03)00044-1
  • Rodgers MR, Blackstone BJ, Reyes AL, Covert TC. Colonisation of point of use water filters by silver resistant non-tuberculous mycobacteria. J Clin Pathol. 1999;52(8):629. doi:10.1136/jcp.52.8.629a
  • Li XZ, Nikaido H, Williams KE. Silver-resistant mutants of Escherichia coli display active efflux of Ag+ and are deficient in porins. J Bacteriol. 1997;179(19):6127–6132. doi:10.1128/JB.179.19.6127-6132.19979324262
  • Gupta A, Matsui K, Lo J-F, Silver S. Molecular basis for resistance to silver cations in Salmonella. Nat Med. 1999;5(2):183–188. doi:10.1038/55459930866
  • Li W, Zhang H, Assaraf YG, et al. Overcoming ABC transporter-mediated multidrug resistance: molecular mechanisms and novel therapeutic drug strategies. Drug Resist Updat Rev Comment Antimicrob Anticancer Chemother. 2016;27:14–29. doi:10.1016/j.drup.2016.05.001