Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1486 word(s) 1486 2021-10-11 09:45:29 |
2 Format Change Meta information modification 1486 2021-10-20 03:25:01 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Gupta, P. Nanomaterials in the Management of Gram-Negative Bacterial Infections. Encyclopedia. Available online: https://encyclopedia.pub/entry/15143 (accessed on 20 April 2024).
Gupta P. Nanomaterials in the Management of Gram-Negative Bacterial Infections. Encyclopedia. Available at: https://encyclopedia.pub/entry/15143. Accessed April 20, 2024.
Gupta, Piyush. "Nanomaterials in the Management of Gram-Negative Bacterial Infections" Encyclopedia, https://encyclopedia.pub/entry/15143 (accessed April 20, 2024).
Gupta, P. (2021, October 19). Nanomaterials in the Management of Gram-Negative Bacterial Infections. In Encyclopedia. https://encyclopedia.pub/entry/15143
Gupta, Piyush. "Nanomaterials in the Management of Gram-Negative Bacterial Infections." Encyclopedia. Web. 19 October, 2021.
Nanomaterials in the Management of Gram-Negative Bacterial Infections
Edit

The exploration of multiplexed bacterial virulence factors is a major problem in the early stages of Escherichia coli infection therapy. Traditional methods for detecting Escherichia coli (E. coli), such as serological experiments, immunoassays, polymerase chain reaction, and isothermal microcalorimetry have some drawbacks. As a result, detecting E. coli in a timely, cost-effective, and sensitive manner is critical for various areas of human safety and health. Intelligent devices based on nanotechnology are paving the way for fast and early detection of E. coli at the point of care. Due to their specific optical, magnetic, and electrical capabilities, nanostructures can play an important role in bacterial sensors. Another one of the applications involved use of nanomaterials in fighting microbial infections, including E. coli mediated infections. Various types of nanomaterials, either used directly as an antibacterial agent such as metallic nanoparticles (NPs) (silver, gold, zinc, etc.), or as a nanocarrier to deliver and target the antibiotic to the E. coli and its infected area. Among different types, polymeric NPs, lipidic nanocarriers, metallic nanocarriers, nanomicelles, nanoemulsion/ nanosuspension, dendrimers, graphene, etc. proved to be effective vehicles to deliver the drug in a controlled fashion at the targeted site with lower off-site drug leakage and side effects.

Escherichia coli nanotechnology infection diagnosis treatment

1. Introduction

Escherichia coli (E. coli) is a gram-negative bacteria and causative agent of many infectious diseases in humans. Many bacterial infections such as urinary tract infections, bloodstream infections, pneumonia, surgical site infections [1][2][3], bacterial sepsis [4][5], and neonatal bacterial meningitis are mainly produced by E. coli [6].
The Gram-negative bacteria are characterized by their cell envelopes, which are composed of a thin peptidoglycan cell wall sandwiched between an inner cytoplasmic cell membrane and a bacterial outer membrane (OM) [7][8]. The OM is an additional protection layer that prevents several substances from entering the bacterium. Nevertheless, OM comprises channels named porins, which allow access to numerous molecules such as drugs [9]. The OM of Gram-negative bacteria is the leading cause of resistance for a wide range of antibiotics such as β-lactams, quinolones, and other antibiotics [10]. Most antibiotics must pass through OM for effective targeting [11]. Hydrophobic molecules can penetrate through the diffusion pathway; in contrast, hydrophilic antibiotics, including β-lactams can pass via porins. Any variation in the OM by Gram-negative bacteria, including mutations in porins, can cause resistance [12].
The use of antibiotics is an efficient, prevailing, and the utmost method for treating E. coli infections. However, huge numbers of drug-resistance strains have appeared due to antibiotics misuse in the last 50 years [13][14]. Furthermore, the inappropriate and overuse of antimicrobial agents has increased pathogens and humans’ resistance [15]. Numerous antibacterial agents such as ampicillin, cotrimoxazole, azithromycin, and gentamicin for E. coli therapy have been revealed to stimulate the Shiga toxin release from E. coli [16]. In addition, the antibodies treatment is an effective method for deactivating the virulence factors and toxins from E. coli [17]. Still, the specificity of antibodies is a major challenge for treating E. coli infections using antibodies [18]. Vaccine therapy using inactivated E. coli has been used to robust the immune responses in humans. However, the short duration of the vaccine producing immunity against bacterial infections is a major drawback for treating E. coli [19]. Despite this, antibiotics-based therapy is still the main strategy against bacterial infections. There is a need to discover new antibacterial agents with new mechanisms to combat resistant bacterial strains [20].
Conventional methods have been used for the diagnosis of E. coli infections for several years, including enzyme-linked immune sorbent assay (ELISA) and polymerase chain reaction (PCR) [21]. The non-culturing approaches are conducted by staining the urine sample for the detection of bacterial infections, but these approaches are time-consuming with less precision value [22]. Meanwhile, the culturing method is one of the oldest techniques for detecting infectious bacteria. Few drawbacks accompany this method, e.g., preparation of individual culture medium to detect each microorganism in the sample for optimal growth [23]. PCR-based methods have been utilized for the identification and diagnosis of bacterial infections [24]. A multiplex PCR test has been established to recognize E. coli producing bacterial infections [25]. ELISA is also one of the molecular techniques widely used to detect bacterial components in the sample [26]. Nevertheless, the prolonged incubation period, extensive sample cleaning, and purification of biomolecules are major disadvantages of these methods [27][28]. To tackle limitations related to the approaches mentioned above, nanotechnology is a quick, efficient and versatile solution for treating and detecting bacterial infections [29]. Recently, numerous NPs, such as silver NPs, zinc oxide NPs, and cationic surfactant NPs, have been used for bacterial infection treatment [30][31][32]. The antibacterial potential of silver NPs generally depends on the particle size, shape and surface modification [33][34][35][36]. The loading of antibacterial moiety into the silver NPs also enhances its antimicrobial activity [37][38]. Zinc oxide is a multifunctional inorganic material that has been used widely in optoelectronic devices, textiles, cosmeceuticals, and most importantly, as an antibacterial agent [39]. The cationic surface NPs are positively charged and can kill bacteria by disrupting bacterial cell wall/membrane, generating free radicals [40][41].
Nanotechnology-based approaches such as gold NPs, silver NPs, magnetic NPs, and quantum dots (QDs) reveal selective target-binding characteristics [42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57]. These characteristics make them ideal candidates for the diagnosis and biosensing of E. coli infections [58][59][60][61][62][63]. The binding to specific ligands such as antibodies and enzymes for detecting bacterial infections is due to the surface properties of NPs. This boosts the specificity of the nanosensor being developed [64]. The entrapment of NPs into nanosensors also enhances the rapid detecting ability of the portable device. NPs, portable devices, nanotubes, nanowires, and nanomechanical devices are typical examples of functional probes for the detection and disinfection of pathogens and other contaminants in different mediums [65][66][67].

2. Point of Care (POC) Devices for Clinical Applications

Pathogens, and all diseases associated with them, are a significant concern worldwide [68]. Diagnostic tests have been suggested to prolong the effectiveness of current antimicrobials; culture and other conventional diagnostics are hindered in their practicality as they are time- and labour-intensive to perform. POC testing is performed near where the patient is being treated and can provide timely results that allow evidence-based clinical interventions to be made (Figure 11) [69]. For example, a portable multiplexed bar-chart SpinChip (MB-SpinChip) integrated with NP-mediated magnetic aptasensors was developed for visual, quantitative instrument-free detection of multiple pathogens. This versatile multiplexed SpinChip combines aptamer-specific recognition and NP-catalysed pressure amplification to achieve a sample-to-answer output for sensitive point-of-care testing (POCT). This user-friendly MB-SpinChip allows visual, quantitative detection of multiple pathogens simultaneously with high sensitivity, but without utilizing any specialized instruments. Using this MB-SpinChip, three major foodborne pathogens, including Salmonella enterica, Escherichia coli, and Listeria monocytogenes, were specifically quantified in apple juice with limits of detection of about 10 CFU/mL [70]. In another study, a smartphone-based nanosensor was developed to detect zika virus (ZIKV) infection. In this light, a nanomotor-based bead-motion cell phone (NBC) system was developed for the immunological detection of ZIKV. The presence of a virus in a testing sample results in the accumulation of platinum (Pt)-nanomotors on the surface of beads, causing their motion in H2O2 solution. Then, the virus concentration is detected in correlation with the change in beads motion. The developed NBC system could detect ZIKV in samples with virus concentrations as low as 1 particle/μL. The NBC platform technology has the potential to be used in the development of point-of-care diagnostics for pathogen detection and disease management in developed and developing countries [71]. Of course, new simulation and machine learning approaches can help better optimize these devices [72][73][74]. The schematic representation of the current analytical methods and POC devices applied for the detection of E. coli are shown in Figure 1.
Figure 1. Schematic representation of the current analytical methods and POC devices applied to detect E. coli Reprinted from ref. [68].

3. Regulatory Landscape of Nanotechnology in Biomedical Applications

The safety assessment of medical devices containing or deriving from nanotechnology is carried out by the US-FDA’s Centre for Devices and Radiological Health (CDRH), housing a Nanotechnology Regulatory Science Research Programme that is based on three pillars: physicochemical characterization methods, in vitro and in vivo models, and (toxicological) risk assessment [75][76]. The types of devices that incorporate nanotechnology include antimicrobial, dental, orthopaedic, neurological, and combination devices and in vitro diagnostic tools. They use various nanomaterials, including silver, zirconia, titanium and titanium dioxide, iron oxides, polymers, gold, graphene etc. Safety assessment of such medical devices should encompass the determination of the rate and magnitude of the nanomaterials into the body for which fit-for-the-purpose in vitro tests would be desirable [77]. Moreover, advanced toxicological risk assessment approaches should support the understanding that the release and patient exposure results in adverse health impacts. It is important to know whether NPs affect the accuracy and/or reliability of standard biocompatibility or toxicity test assays, such as cytotoxicity and genotoxicity. Because of the vast number of sizes, shapes, and chemistry of nanomaterials, there is the need for the development of in vitro models (2D, 3D, organ on a chip, organoids) and in silico models in order to predict human responses and improve in vitro to in vivo extrapolations [76].

References

  1. Weiner, L.M.; Webb, A.K.; Limbago, B.; Dudeck, M.A.; Patel, J.; Kallen, A.J.; Edwards, J.R.; Sievert, D.M. Antimicrobial-resistant pathogens associated with healthcare-associated infections: Summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2011–2014. Infect. Control. Hosp. Epidemiol. 2016, 37, 1288–1301.
  2. Al-Hasan, M.N.; Lahr, B.D.; Eckel-Passow, J.E.; Baddour, L.M. Antimicrobial resistance trends of Escherichia coli bloodstream isolates: A population-based study, 1998–2007. J. Antimicrob. Chemother. 2009, 64, 169–174.
  3. Søgaard, M.; Nørgaard, M.; Dethlefsen, C.; Schønheyder, H.C. Temporal changes in the incidence and 30-day mortality associated with bacteremia in hospitalized patients from 1992 through 2006: A population-based cohort study. Clin. Infect. Dis. 2011, 52, 61–69.
  4. Tavafi, H.; Sadrzadeh-Afshar, M.; Niroomand, S. In Vitro Effectiveness of Antimicrobial Properties of Propolis and Chlorhexidine on Oral Pathogens: A Comparative Study: Effectiveness of Antimicrobial Properties of Propolis and Chlorhexidine on Oral Pathogens. Biosis Biol. Syst. 2020, 1, 116–125.
  5. Opal, S.M.; Garber, G.E.; LaRosa, S.P.; Maki, D.G.; Freebairn, R.C.; Kinasewitz, G.T.; Dhainaut, J.-F.; Yan, S.B.; Williams, M.D.; Graham, D.E. Systemic host responses in severe sepsis analyzed by causative microorganism and treatment effects of drotrecogin alfa (activated). Clin. Infect. Dis. 2003, 37, 50–58.
  6. Ranieri, V.M.; Thompson, B.T.; Barie, P.S.; Dhainaut, J.-F.; Douglas, I.S.; Finfer, S.; Gårdlund, B.; Marshall, J.C.; Rhodes, A.; Artigas, A. Drotrecogin alfa (activated) in adults with septic shock. N. Engl. J. Med. 2012, 366, 2055–2064.
  7. Hauser, A. Cell Envelope. Antibiotic Basic for Clinicians; Wolters Kluwer India Pvt Ltd.: Gurugram, India, 2015.
  8. Dasaraju, P.V.; Liu, C. Infections of the respiratory system. In Medical Microbiology, 4th ed.; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996.
  9. Kapoor, G.; Saigal, S.; Elongavan, A. Action and resistance mechanisms of antibiotics: A guide for clinicians. J. Anaesthesiol. Clin. Pharmacol. 2017, 33, 300.
  10. Pang, X.; Gong, K.; Zhang, X.; Wu, S.; Cui, Y.; Qian, B.-Z. Osteopontin as a multifaceted driver of bone metastasis and drug resistance. Pharmacol. Res. 2019, 144, 235–244.
  11. Miller, S.I. Antibiotic resistance and regulation of the gram-negative bacterial outer membrane barrier by host innate immune molecules. MBio 2016, 7, e01541-16.
  12. Gupta, V.; Datta, P. Next-generation strategy for treating drug resistant bacteria: Antibiotic hybrids. Indian J. Med. Res. 2019, 149, 97.
  13. Pandey, V.K.; Srivastava, K.R.; Ajmal, G.; Thakur, V.K.; Gupta, V.K.; Upadhyay, S.N.; Mishra, P.K. Differential Susceptibility of Catheter Biomaterials to Biofilm-Associated Infections and Their Remedy by Drug-Encapsulated Eudragit RL100 Nanoparticles. Int. J. Mol. Sci. 2019, 20, 5110.
  14. Bansal, K.K.; Bhardwaj, J.K.; Saraf, P.; Thakur, V.K.; Sharma, P.C. Synthesis of Thiazole Clubbed Pyrazole Derivatives as Apoptosis Inducers and Anti-Infective Agents. Mater. Today Chem. 2020, 17, 100335.
  15. Tavafi, H. An Investigation of Antibacterial Resistance Patterns in Isolated Bacteria from Contaminated Water Samples in Poultry Slaughterhouses. Biosis Biol. Syst. 2020, 1, 85–90.
  16. Mohsin, M.; Haque, A.; Ali, A.; Sarwar, Y.; Bashir, S.; Tariq, A.; Afzal, A.; Iftikhar, T.; Saeed, M.A. Effects of ampicillin, gentamicin, and cefotaxime on the release of Shiga toxins from Shiga toxin–producing Escherichia coli isolated during a diarrhea episode in Faisalabad, Pakistan. Foodborne Pathog. Dis. 2010, 7, 85–90.
  17. Cheng, L.W.; Henderson, T.D.; Patfield, S.; Stanker, L.H.; He, X. Mouse in vivo neutralization of Escherichia coli Shiga toxin 2 with monoclonal antibodies. Toxins 2013, 5, 1845–1858.
  18. Berry, J.D.; Gaudet, R.G. Antibodies in infectious diseases: Polyclonals, monoclonals and niche biotechnology. New Biotechnol. 2011, 28, 489–501.
  19. Saeedi, P.; Yazdanparast, M.; Behzadi, E.; Salmanian, A.H.; Mousavi, S.L.; Nazarian, S.; Amani, J. A review on strategies for decreasing E. coli O157: H7 risk in animals. Microb. Pathog. 2017, 103, 186–195.
  20. Ling, L.L.; Schneider, T.; Peoples, A.J.; Spoering, A.L.; Engels, I.; Conlon, B.P.; Mueller, A.; Schäberle, T.F.; Hughes, D.E.; Epstein, S. A new antibiotic kills pathogens without detectable resistance. Nature 2015, 517, 455–459.
  21. Kumar, M.; Ghosh, S.; Nayak, S.; Das, A. Recent advances in biosensor based diagnosis of urinary tract infection. Biosens. Bioelectron. 2016, 80, 497–510.
  22. Van Nostrand, J.D.; Junkins, A.D.; Bartholdi, R.K. Poor predictive ability of urinalysis and microscopic examination to detect urinary tract infection. Am. J. Clin. Pathol. 2000, 113, 709–713.
  23. Graham, J.; Galloway, A. ACP Best Practice No 167: The laboratory diagnosis of urinary tract infection. J. Clin. Pathol. 2001, 54, 911–919.
  24. Kubista, M.; Andrade, J.M.; Bengtsson, M.; Forootan, A.; Jonák, J.; Lind, K.; Sindelka, R.; Sjöback, R.; Sjögreen, B.; Strömbom, L. The real-time polymerase chain reaction. Mol. Asp. Med. 2006, 27, 95–125.
  25. Padmavathy, á.; Kumar, R.V.; Patel, A.; Swarnam, S.D.; Vaidehi, T.; Ali, B.J. Rapid and sensitive detection of major uropathogens in a single-pot multiplex PCR assay. Curr. Microbiol. 2012, 65, 44–53.
  26. Mohanan, P.V.; Banerjee, S.; Geetha, C.S. Detection of pyrogenicity on medical grade polymer materials using rabbit pyrogen, LAL and ELISA method. J. Pharm. Biomed. Anal. 2011, 55, 1170–1174.
  27. Ivnitski, D.; Abdel-Hamid, I.; Atanasov, P.; Wilkins, E. Biosensors for detection of pathogenic bacteria. Biosens. Bioelectron. 1999, 14, 599–624.
  28. Skottrup, P.D.; Nicolaisen, M.; Justesen, A.F. Towards on-site pathogen detection using antibody-based sensors. Biosens. Bioelectron. 2008, 24, 339–348.
  29. Carvalho, F.; George, J.; Sheikh, H.M.A.; Selvin, R. Advances in screening, detection and enumeration of Escherichia coli using nanotechnology-based methods: A review. J. Biomed. Nanotechnol. 2018, 14, 829–846.
  30. Ates, B.; Koytepe, S.; Ulu, A.; Gurses, C.; Thakur, V.K. Chemistry, Structures, and Advanced Applications of Nanocomposites from Biorenewable Resources. Chem. Rev. 2020, 120, 9304–9362.
  31. Morsy, M.K.; Khalaf, H.H.; Sharoba, A.M.; El-Tanahi, H.H.; Cutter, C.N. Incorporation of essential oils and nanoparticles in pullulan films to control foodborne pathogens on meat and poultry products. J. Food Sci. 2014, 79, M675–M684.
  32. Paredes, D.; Ortiz, C.; Torres, R. Synthesis, characterization, and evaluation of antibacterial effect of Ag nanoparticles against Escherichia coli O157: H7 and methicillin-resistant Staphylococcus aureus (MRSA). Int. J. Nanomed. 2014, 9, 1717.
  33. Smekalova, M.; Aragon, V.; Panacek, A.; Prucek, R.; Zboril, R.; Kvitek, L. Enhanced antibacterial effect of antibiotics in combination with silver nanoparticles against animal pathogens. Vet. J. 2016, 209, 174–179.
  34. Hari, N.; Thomas, T.K.; Nair, A.J. Comparative Study on the Synergistic Action of Garlic Synthesized and Citrate Capped Silver Nanoparticles with β-Penem Antibiotics. Int. Sch. Res. Not. 2013, 2013, 792105.
  35. Fang, J.; Zhong, C.; Mu, R. The study of deposited silver particulate films by simple method for efficient SERS. Chem. Phys. Lett. 2005, 401, 271–275.
  36. Abdel-Azeem, A.; Nada, A.A.; O’donovan, A.; Thakur, V.K.; Elkelish, A. Mycogenic Silver Nanoparticles From Endophytic Trichoderma Atroviride with Antimicrobial Activity. J. Renew. Mater. 2020, 8, 171–185.
  37. Shakeri, S.; Ashrafizadeh, M.; Zarrabi, A.; Roghanian, R.; Afshar, E.G.; Pardakhty, A.; Mohammadinejad, R.; Kumar, A.; Thakur, V.K. Multifunctional Polymeric Nanoplatforms for Brain Diseases Diagnosis, Therapy and Theranostics. Biomedicines 2020, 8, 13.
  38. Li, P.; Li, J.; Wu, C.; Wu, Q.; Li, J. Synergistic antibacterial effects of β-lactam antibiotic combined with silver nanoparticles. Nanotechnology 2005, 16, 1912.
  39. Oprea, O.; Andronescu, E.; Ficai, D.; Ficai, A.; N Oktar, F.; Yetmez, M. ZnO applications and challenges. Curr. Org. Chem. 2014, 18, 192–203.
  40. Yang, S.-C.; Aljuffali, I.A.; Sung, C.T.; Lin, C.-F.; Fang, J.-Y. Antimicrobial activity of topically-applied soyaethyl morpholinium ethosulfate micelles against Staphylococcus species. Nanomedicine 2016, 11, 657–671.
  41. Pan, D.; Xia, X.-X.; Zhou, H.; Jin, S.-Q.; Lu, Y.-Y.; Liu, H.; Gao, M.-L.; Jin, Z.-B. COCO enhances the efficiency of photoreceptor precursor differentiation in early human embryonic stem cell-derived retinal organoids. Stem Cell Res. Ther. 2020, 11, 366.
  42. Sharma, D.; Shandilya, P.; Saini, N.K.; Singh, P.; Thakur, V.K.; Saini, R.V.; Mittal, D.; Chandan, G.; Saini, V.; Saini, A.K. Insights into the Synthesis and Mechanism of Green Synthesized Antimicrobial Nanoparticles, Answer to the Multidrug Resistance. Mater. Today Chem. 2021, 19, 100391.
  43. Sudhaik, A.; Raizada, P.; Thakur, S.; Saini, R.V.; Saini, A.K.; Singh, P.; Kumar Thakur, V.; Nguyen, V.-H.; Khan, A.A.P.; Asiri, A.M. Synergistic Photocatalytic Mitigation of Imidacloprid Pesticide and Antibacterial Activity Using Carbon Nanotube Decorated Phosphorus Doped Graphitic Carbon Nitride Photocatalyst. J. Taiwan Inst. Chem. Eng. 2020, 113, 142–154.
  44. Nazaripour, E.; Mousazadeh, F.; Moghadam, M.D.; Najafi, K.; Borhani, F.; Sarani, M.; Ghasemi, M.; Rahdar, A.; Iravani, S.; Khatami, M. Biosynthesis of lead oxide and cerium oxide nanoparticles and their cytotoxic activities against colon cancer cell line. Inorg. Chem. Commun. 2021, 131, 108800.
  45. M Balasubramaniam, B.; Prateek; Ranjan, S.; Saraf, M.; Kar, P.; Singh, S.P.; Thakur, V.K.; Singh, A.; Gupta, R.K. Antibacterial and Antiviral Functional Materials: Chemistry and Biological Activity toward Tackling COVID-19-like Pandemics. ACS Pharmacol. Transl. Sci. 2021, 4, 8–54.
  46. Siwal, S.S.; Zhang, Q.; Saini, A.K.; Thakur, V.K. Antimicrobial Materials: New Strategies to Tackle Various Pandemics. J. Renew. Mater. 2020, 8, 1543–1563.
  47. Arshad, R.; Pal, K.; Sabir, F.; Rahdar, A.; Bilal, M.; Shahnaz, G.; Kyzas, G.Z. A review of the nanomaterials use for the diagnosis and therapy of salmonella typhi. J. Mol. Struct. 2021, 1230, 129928.
  48. Hakami, T.M.; Davarpanah, A.; Rahdar, A.; Barrett, S. Structural and magnetic study and cytotoxicity evaluation of tetra-metallic nanoparticles of Co0. 5Ni0. 5CrxFe2-xO4 prepared by co-precipitation. J. Mol. Struct. 2018, 1165, 344–348.
  49. Hasanein, P.; Rahdar, A.; Bahabadi, S.E.; Kumar, A.; Kyzas, G.Z. Manganese/cerium nanoferrites: Synthesis and toxicological effects by intraperitoneal administration in rats. Inorg. Chem. Commun. 2021, 125, 108433.
  50. Mohammadi, L.; Pal, K.; Bilal, M.; Rahdar, A.; Fytianos, G.; Kyzas, G.Z. Green nanoparticles to treat patients from Malaria disease: An overview. J. Mol. Struct. 2021, 1299, 129857.
  51. Nikazar, S.; Sivasankarapillai, V.S.; Rahdar, A.; Gasmi, S.; Anumol, P.; Shanavas, M.S. Revisiting the cytotoxicity of quantum dots: An in-depth overview. Biophys. Rev. 2020, 12, 703–718.
  52. Pillai, A.M.; Sivasankarapillai, V.S.; Rahdar, A.; Joseph, J.; Sadeghfar, F.; Rajesh, K.; Kyzas, G.Z. Green synthesis and characterization of zinc oxide nanoparticles with antibacterial and antifungal activity. J. Mol. Struct. 2020, 1211, 128107.
  53. Rahdar, A.; Aliahmad, M.; Samani, M.; HeidariMajd, M.; Susan, M.A.B.H. Synthesis and characterization of highly efficacious Fe-doped ceria nanoparticles for cytotoxic and antifungal activity. Ceram. Int. 2019, 45, 7950–7955.
  54. Rahdar, A.; Beyzaei, H.; Askari, F.; Kyzas, G.Z. Gum-based cerium oxide nanoparticles for antimicrobial assay. Appl. Phys. A 2020, 126, 324.
  55. Rahdar, A.; Hajinezhad, M.R.; Hamishekar, H.; Ghamkhari, A.; Kyzas, G.Z. Copolymer/graphene oxide nanocomposites as potential anticancer agents. Polym. Bull. 2020, 78, 4877–4898.
  56. Rahdar, A.; Taboada, P.; Aliahmad, M.; Hajinezhad, M.R.; Sadeghfar, F. Iron oxide nanoparticles: Synthesis, physical characterization, and intraperitoneal biochemical studies in Rattus norvegicus. J. Mol. Struct. 2018, 1173, 240–245.
  57. Taimoory, S.M.; Rahdar, A.; Aliahmad, M.; Sadeghfar, F.; Hajinezhad, M.R.; Jahantigh, M.; Shahbazi, P.; Trant, J.F. The synthesis and characterization of a magnetite nanoparticle with potent antibacterial activity and low mammalian toxicity. J. Mol. Liq. 2018, 265, 96–104.
  58. Naravaneni, R.; Jamil, K. Rapid detection of food-borne pathogens by using molecular techniques. J. Med. Microbiol. 2005, 54, 51–54.
  59. Mukherjee, S.; Sau, S.; Madhuri, D.; Bollu, V.S.; Madhusudana, K.; Sreedhar, B.; Banerjee, R.; Patra, C.R. Green synthesis and characterization of monodispersed gold nanoparticles: Toxicity study, delivery of doxorubicin and its bio-distribution in mouse model. J. Biomed. Nanotechnol. 2016, 12, 165–181.
  60. Bhatt, K.D.; Vyas, D.J.; Makwana, B.A.; Darjee, S.M.; Jain, V.K.; Shah, H. Turn-on fluorescence probe for selective detection of Hg (II) by calixpyrrole hydrazide reduced silver nanoparticle: Application to real water sample. Chin. Chem. Lett. 2016, 27, 731–737.
  61. Zou, Q.; Xing, P.; Wei, L.; Liu, B. Gene2vec: Gene subsequence embedding for prediction of mammalian N6-methyladenosine sites from mRNA. RNA 2019, 25, 205–218.
  62. Deng, X.; Xu, T.; Huang, G.; Li, Q.; Luo, L.; Zhao, Y.; Wu, Z.; Ou-Yang, J.; Yang, X.; Xie, M. Design and Fabrication of a Novel Dual-Frequency Confocal Ultrasound Transducer for Microvessels Super-Harmonic Imaging. IEEE Trans. Ultrason. Ferroelectr. Freq. Control 2020, 68, 1272–1277.
  63. Badoei-Dalfard, A.; Sohrabi, N.; Karami, Z.; Sargazi, G. Fabrication of an efficient and sensitive colorimetric biosensor based on Uricase/Th-MOF for uric acid sensing in biological samples. Biosens. Bioelectron. 2019, 141, 111420.
  64. Heo, J.; Hua, S.Z. An overview of recent strategies in pathogen sensing. Sensors 2009, 9, 4483–4502.
  65. Nugen, S.; Baeumner, A. Trends and opportunities in food pathogen detection. Anal. Bioanal. Chem. 2008, 391, 451–454.
  66. Kumar, R.; Raizada, P.; Verma, N.; Hosseini-Bandegharaei, A.; Thakur, V.K.; Le, Q.V.; Nguyen, V.-H.; Selvasembian, R.; Singh, P. Recent Advances on Water Disinfection Using Bismuth Based Modified Photocatalysts: Strategies and Challenges. J. Clean. Prod. 2021, 297, 126617.
  67. Sharma, P.C.; Sharma, D.; Sharma, A.; Saini, N.; Goyal, R.; Ola, M.; Chawla, R.; Thakur, V.K. Hydrazone Comprising Compounds as Promising Anti-Infective Agents: Chemistry and Structure-Property Relationship. Mater. Today Chem. 2020, 18, 100349.
  68. Canciu, A.; Tertis, M.; Hosu, O.; Cernat, A.; Cristea, C.; Graur, F. Modern analytical techniques for detection of bacteria in surface and wastewaters. Sustainability 2021, 13, 7229.
  69. Reali, S.; Najib, E.Y.; Balázs, K.E.T.; Tan, A.C.H.; Váradi, L.; Hibbs, D.E.; Groundwater, P.W. Novel diagnostics for point-of-care bacterial detection and identification. RSC Adv. 2019, 9, 21486–21497.
  70. Wei, X.; Zhou, W.; Sanjay, S.T.; Zhang, J.; Jin, Q.; Xu, F.; Dominguez, D.C.; Li, X. Multiplexed instrument-free bar-chart spinchip integrated with nanoparticle-mediated magnetic aptasensors for visual quantitative detection of multiple pathogens. Anal. Chem. 2018, 90, 9888–9896.
  71. Draz, M.S.; Lakshminaraasimulu, N.K.; Krishnakumar, S.; Battalapalli, D.; Vasan, A.; Kanakasabapathy, M.K.; Sreeram, A.; Kallakuri, S.; Thirumalaraju, P.; Li, Y. Motion-based immunological detection of Zika virus using Pt-nanomotors and a cellphone. ACS Nano 2018, 12, 5709–5718.
  72. Wang, X.-F.; Gao, P.; Liu, Y.-F.; Li, H.-F.; Lu, F. Predicting thermophilic proteins by machine learning. Curr. Bioinform. 2020, 15, 493–502.
  73. Niu, M.; Lin, Y.; Zou, Q. sgRNACNN: Identifying sgRNA on-target activity in four crops using ensembles of convolutional neural networks. Plant Mol. Biol. 2021, 105, 483–495.
  74. Sun, S.; Xu, L.; Zou, Q.; Wang, G. BP4RNAseq: A babysitter package for retrospective and newly generated RNA-seq data analyses using both alignment-based and alignment-free quantification method. Bioinformatics 2021, 37, 1319–1321.
  75. Bowman, D.M.; Ludlow, K. Assessing the impact of a for government review on the nanotechnology regulatory landscape. Monash UL Rev. 2012, 38, 168.
  76. Allan, J.; Belz, S.; Hoeveler, A.; Hugas, M.; Okuda, H.; Patri, A.; Rauscher, H.; Silva, P.; Slikker, W.; Sokull-Kluettgen, B. Regulatory landscape of nanotechnology and nanoplastics from a global perspective. Regul. Toxicol. Pharmacol. 2021, 122, 104885.
  77. Mirza, M.A.; Iqbal, Z.; Mishra, H. FDC in nanotechnology: Regulatory landscape. In Nanocarriers for the Delivery of Combination Drugs; Elsevier: Amsterdam, The Netherlands, 2021; pp. 473–496.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 467
Revisions: 2 times (View History)
Update Date: 20 Oct 2021
1000/1000