151
|
Brebels J, Mignon A. Polymer-Based Constructs for Flexor Tendon Repair: A Review. Polymers (Basel) 2022; 14:867. [PMID: 35267690 PMCID: PMC8912457 DOI: 10.3390/polym14050867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 02/04/2023] Open
Abstract
A flexor tendon injury is acquired fast and is common for athletes, construction workers, and military personnel among others, treated in the emergency department. However, the healing of injured flexor tendons is stretched over a long period of up to 12 weeks, therefore, remaining a significant clinical problem. Postoperative complications, arising after traditional tendon repair strategies, include adhesion and tendon scar tissue formation, insufficient mechanical strength for early active mobilization, and infections. Various researchers have tried to develop innovative strategies for developing a polymer-based construct that minimalizes these postoperative complications, yet none are routinely used in clinical practice. Understanding the role such constructs play in tendon repair should enable a more targeted approach. This review mainly describes the polymer-based constructs that show promising results in solving these complications, in the hope that one day these will be used as a routine practice in flexor tendon repair, increasing the well-being of the patients. In addition, the review also focuses on the incorporation of active compounds in these constructs, to provide an enhanced healing environment for the flexor tendon.
Collapse
Affiliation(s)
| | - Arn Mignon
- Surface and Interface Engineered Materials, Campus Group T, KU Leuven, Andreas Vesaliusstraat 13, 3000 Leuven, Belgium;
| |
Collapse
|
152
|
Tong Q, Li T, Jiang L, Wang Z, Qian Y. Nanoparticle, a promising therapeutic strategy for the treatment of infective endocarditis. Anatol J Cardiol 2022; 26:90-99. [PMID: 35190356 PMCID: PMC8878918 DOI: 10.5152/anatoljcardiol.2021.867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 07/30/2023] Open
Abstract
Infective endocarditis (IE) has been recognized as a biofilm-related disease caused by pathogenic microorganisms, such as bacteria and fungi that invade and damage the heart valves and endocardium. There are many difficulties and challenges in the antimicrobial treatment of IE, including multi-drug resistant pathogens, large dose of drug administration with following side effects, and poor prognosis. For the past few years, the development of nanotechnology has promoted the use of nanoparticles as antimicrobial nano-pharmaceuticals or novel drug delivery systems (NDDS) in antimicrobial therapy for chronic infections and biofilm-related infectious disease as these molecules exhibit several advantages. Therefore, nanoparticles have a potential role to play in solving problems in the treatment of IE, including improving antimicrobial activity, increasing drug bioavailability, minimizing frequency of drug administration, and preventing side effects. In this article, we review the latest advances in nanoparticles against drug-resistant bacteria in biofilm and recommends nanoparticles as an alternative strategy to the antibiotic treatment of IE.
Collapse
Affiliation(s)
- Qi Tong
- Department of Cardiovascular Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University; Chengdu-China
| | - Tao Li
- Department of Cardiovascular Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University; Chengdu-China
| | - Lu Jiang
- Department of Cardiovascular Surgery, Sichuan Provincial People's University of Electronic Science and Technology of China; Chengdu-China
| | - Zhengjie Wang
- Department of Cardiovascular Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University; Chengdu-China
| | - Yongjun Qian
- Department of Cardiovascular Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University; Chengdu-China
| |
Collapse
|
153
|
Lai CKC, Ng RWY, Leung SSY, Hui M, Ip M. Overcoming the rising incidence and evolving mechanisms of antibiotic resistance by novel drug delivery approaches - An overview. Adv Drug Deliv Rev 2022; 181:114078. [PMID: 34896131 DOI: 10.1016/j.addr.2021.114078] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/14/2022]
Abstract
Antimicrobial resistance is a normal evolutionary process for microorganisms. Antibiotics exerted accelerated selective pressure that hasten bacterial resistance through mutation, and acquisition external genes. These genes often carry multiple antibiotic resistant determinants allowing the recipient microbe an instant "super-bug" status. The extent of Antimicrobial Resistance (AMR) has reached a level of global crisis, existing antimicrobials are no long effective in treating infections caused by AMR pathogens. The great majority of clinically available antimicrobial agents are administered through oral and intra-venous routes. Overcoming antibacterial resistance by novel drug delivery approach offered new hopes, particularly in the treatment of AMR pathogens in sites less assessible through systemic circulation such as the lung and skin. In the current review, we will revisit the mechanism and incidence of important AMR pathogens. Finally, we will discuss novel drug delivery approaches including novel local antibiotic delivery systems, hybrid antibiotics, and nanoparticle-based antibiotic delivery systems.
Collapse
Affiliation(s)
- Christopher K C Lai
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| | - Rita W Y Ng
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| | - Sharon S Y Leung
- School of Pharmacy, The Chinese University of Hong Kong, New Territories, Hong Kong Special Administrative Region.
| | - Mamie Hui
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| | - Margaret Ip
- Department of Microbiology, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong Special Administrative Region.
| |
Collapse
|
154
|
Hwang J, Mros S, Gamble AB, Tyndall JDA, McDowell A. Improving Antibacterial Activity of a HtrA Protease Inhibitor JO146 against Helicobacter pylori: A Novel Approach Using Microfluidics-Engineered PLGA Nanoparticles. Pharmaceutics 2022; 14:pharmaceutics14020348. [PMID: 35214080 PMCID: PMC8875321 DOI: 10.3390/pharmaceutics14020348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/24/2022] [Accepted: 01/29/2022] [Indexed: 11/16/2022] Open
Abstract
Nanoparticle drug delivery systems have emerged as a promising strategy for overcoming limitations of antimicrobial drugs such as stability, bioavailability, and insufficient exposure to the hard-to-reach bacterial drug targets. Although size is a vital colloidal feature of nanoparticles that governs biological interactions, the absence of well-defined size control technology has hampered the investigation of optimal nanoparticle size for targeting bacterial cells. Previously, we identified a lead antichlamydial compound JO146 against the high temperature requirement A (HtrA) protease, a promising antibacterial target involved in protein quality control and virulence. Here, we reveal that JO146 was active against Helicobacter pylori with a minimum bactericidal concentration of 18.8–75.2 µg/mL. Microfluidic technology using a design of experiments approach was utilized to formulate JO146-loaded poly(lactic-co-glycolic) acid nanoparticles and explore the effect of the nanoparticle size on drug delivery. JO146-loaded nanoparticles of three different sizes (90, 150, and 220 nm) were formulated with uniform particle size distribution and drug encapsulation efficiency of up to 25%. In in vitro microdilution inhibition assays, 90 nm nanoparticles improved the minimum bactericidal concentration of JO146 two-fold against H. pylori compared to the free drug alone, highlighting that controlled engineering of nanoparticle size is important in drug delivery optimization.
Collapse
Affiliation(s)
- Jimin Hwang
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand; (J.H.); (A.B.G.); (J.D.A.T.)
| | - Sonya Mros
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand;
| | - Allan B. Gamble
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand; (J.H.); (A.B.G.); (J.D.A.T.)
| | - Joel D. A. Tyndall
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand; (J.H.); (A.B.G.); (J.D.A.T.)
| | - Arlene McDowell
- School of Pharmacy, University of Otago, Dunedin 9054, New Zealand; (J.H.); (A.B.G.); (J.D.A.T.)
- Correspondence:
| |
Collapse
|
155
|
Chen Y, Huang Y, Jin Q. Polymeric nanoplatforms for the delivery of antibacterial agents. MACROMOL CHEM PHYS 2022. [DOI: 10.1002/macp.202100440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yongcheng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education Department of Polymer Science and Engineering Zhejiang University Hangzhou Zhejiang 310027 PR China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education Department of Polymer Science and Engineering Zhejiang University Hangzhou Zhejiang 310027 PR China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education Department of Polymer Science and Engineering Zhejiang University Hangzhou Zhejiang 310027 PR China
| |
Collapse
|
156
|
Metelkina O, Huck B, O'Connor JS, Koch M, Manz A, Lehr CM, Titz A. Targeting extracellular lectins of Pseudomonas aeruginosa with glycomimetic liposomes. J Mater Chem B 2022; 10:537-548. [PMID: 34985094 DOI: 10.1039/d1tb02086b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The antimicrobial resistance crisis requires novel approaches for the therapy of infections especially with Gram-negative pathogens. Pseudomonas aeruginosa is defined as priority 1 pathogen by the WHO and thus of particular interest. Its drug resistance is primarily associated with biofilm formation and essential constituents of its extracellular biofilm matrix are the two lectins, LecA and LecB. Here, we report microbial lectin-specific targeted nanovehicles based on liposomes. LecA- and LecB-targeted phospholipids were synthesized and used for the preparation of liposomes. These liposomes with varying surface ligand density were then analyzed for their competitive and direct lectin binding activity. We have further developed a microfluidic device that allowed the optical detection of the targeting process to the bacterial lectins. Our data showed that the targeted liposomes are specifically binding to their respective lectin and remain firmly attached to surfaces containing these lectins. This synthetic and biophysical study provides the basis for future application in targeted antibiotic delivery to overcome antimicrobial resistance.
Collapse
Affiliation(s)
- Olga Metelkina
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany. .,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124 Braunschweig, Germany.,Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
| | - Benedikt Huck
- Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Jonathan S O'Connor
- KIST Europe, 66123 Saarbrücken, Germany.,Department of Systems Engineering, Saarland University, 66123 Saarbrücken, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Andreas Manz
- KIST Europe, 66123 Saarbrücken, Germany.,Department of Systems Engineering, Saarland University, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany. .,Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, 38124 Braunschweig, Germany.,Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
157
|
Shein AMS, Hongsing P, Abe S, Luk-In S, Ragupathi NKD, Wannigama DL, Chatsuwan T. Will There Ever Be Cure for Chronic, Life-Changing Colistin-Resistant Klebsiella pneumoniae in Urinary Tract Infection? Front Med (Lausanne) 2022; 8:806849. [PMID: 35004783 PMCID: PMC8740227 DOI: 10.3389/fmed.2021.806849] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/29/2021] [Indexed: 11/21/2022] Open
Affiliation(s)
- Aye Mya Sithu Shein
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.,Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Interdisciplinary Program of Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Parichart Hongsing
- Mae Fah Luang University Hospital, Chiang Rai, Thailand.,School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Shuichi Abe
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Sirirat Luk-In
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Naveen Kumar Devanga Ragupathi
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, United Kingdom.,Department of Chemical and Biological Engineering, The University of Sheffield, Sheffield, United Kingdom.,Department of Clinical Microbiology, Christian Medical College, Vellore, India
| | - Dhammika Leshan Wannigama
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.,Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, United Kingdom.,School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.,Antimicrobial Resistance and Stewardship Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
158
|
Tuchin VV, Genina EA, Tuchina ES, Svetlakova AV, Svenskaya YI. Optical clearing of tissues: Issues of antimicrobial phototherapy and drug delivery. Adv Drug Deliv Rev 2022; 180:114037. [PMID: 34752842 DOI: 10.1016/j.addr.2021.114037] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/23/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
This review presents principles and novelties in the field of tissue optical clearing (TOC) technology, as well as application for optical monitoring of drug delivery and effective antimicrobial phototherapy. TOC is based on altering the optical properties of tissue through the introduction of immersion optical cleaning agents (OCA), which impregnate the tissue of interest. We also analyze various methods and kinetics of delivery of photodynamic agents, nanoantibiotics and their mixtures with OCAs into the tissue depth in the context of antimicrobial and antifungal phototherapy. In vitro and in vivo studies of antimicrobial phototherapies, such as photodynamic, photothermal plasmonic and photocatalytic, are summarized, and the prospects of a new TOC technology for effective killing of pathogens are discussed.
Collapse
|
159
|
Nano-Bio Interactions in the Lung. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_14-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
160
|
Spread of ESβL-producing Escherichia coli and the anti-virulence effect of graphene nano-sheets. Arch Microbiol 2021; 204:51. [DOI: 10.1007/s00203-021-02687-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 10/19/2022]
|
161
|
Wu JF, Hong XD, Jin J, Fei YHH, Zhang MY, Si TT, Fan H, Zhang XD. [Effects of N-trimethyl chitosan-recombinant tissue factor pathway inhibitor complex on avulsion flap with roll compaction in rat]. ZHONGHUA SHAO SHANG ZA ZHI = ZHONGHUA SHAOSHANG ZAZHI = CHINESE JOURNAL OF BURNS 2021; 37:1158-1165. [PMID: 34839594 PMCID: PMC11917342 DOI: 10.3760/cma.j.cn501120-20200914-00409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Objective: To investigate the effect of N-trimethyl chitosan-recombinant tissue factor pathway inhibitor (rTFPI) complex on avulsion flap with roll compaction in rat. Methods: The experimental methods were adopted. The N-trimethyl chitosan-rTFPI complex solution was prepared by ion cross-linking method. The morphology of the complex was observed by scanning electron microscope, and its diameter was measured. The encapsulation rate of rTFPI in the complex and drug loading rate of the complex was determined and calculated by enzyme-linked immunosorbent assay (ELISA) method (n=3). The concentration of rTFPI in the solution at 0, 10, 30, 45, 60, 90, 120, 240 minutes of storage was measured by ELISA method to observe the release of rTFPI, and its half-life was calculated (n=3). Twenty-four 6-week-old male Sprague-Dawley rats were divided into phosphate buffered saline (PBS) group, N-trimethyl chitosan alone group, rTFPI alone group, and N-trimethyl chitosan-rTFPI group according to the random number table, with 6 rats in each group. The avulsion flaps with roll compaction were prepared on the backs of rats with pedicles located on the line of the bilateral iliac spine and lifted from the surface of the muscle membrane. One injection of corresponding reagents was carried out immediately after in-situ suture and on post operation day (POD) 1, 2, and 3. General changes of the flap were observed on POD 1, 3, and 7. On POD 7, the survival area of the flap was measured and the survival rate of the flap was calculated; the flaps were divided into pedicle, proximal, middle, and distal segments, and the blood perfusion in the proximal, middle, and distal segment tissue of the flap was detected by the laser speckle blood flow imager; tissue samples in the middle of the flap were cut and stained with hematoxylin and eosin to observe the changes in tissue structure and the infiltration of inflammatory cells, and the numbers of embolized blood vessels and new blood vessels per 100 times visual field were counted. Data were statistically analyzed with one-way analysis of variance and least significant difference test. Results: The N-trimethyl chitosan-rTFPI complex had an irregular spherical structure with a diameter of 150-200 nm. The encapsulation rate of rTFPI in the complex and drug loading rate of the complex were (88.7±2.1)% and (2.83±0.09)%, respectively. The concentration of rTFPI in the solution of the N-trimethyl chitosan-rTFPI complex gradually increased with prolonged storage time, and the release was basically stable at 90 min, with half-life of (651±36) min. On POD 1, the distal parts of flaps of rats in N-trimethyl chitosan alone group darkened significantly. On POD 3, scabs and necrosis were relatively mild on the distal segment of the flaps of rats in rTFPI alone group and N-trimethyl chitosan-rTFPI group as compared with those of the other two groups. On POD 7, the necrosis boundaries of the flaps of rats in each group were clear. On POD 7, the flap survival rates of rats in rTFPI alone group and N-trimethyl chitosan-rTFPI group were (63±7)% and (73±5)%, respectively, which were significantly higher than (41±3)% in PBS group and (52±7)% in N-trimethyl chitosan alone group. Moreover, the flap survival rate of rats in N-trimethyl chitosan-rTFPI group was significantly higher than that in rTFPI alone group (P<0.05). On POD 7, the flaps of rats in each group had blood perfusion; the blood perfusion values in the proximal segment tissue of the rat flaps in N-trimethyl chitosan alone group and the blood perfusion values in the proximal, middle, and distal segment tissue of the rat flaps in rTFPI alone group and N-trimethyl chitosan-rTFPI group were significantly higher than those in PBS group (P<0.05 or P<0.01); the blood perfusion values in the distal segment tissue of the rat flaps in rTFPI alone group and the blood perfusion values in the middle and distal segment tissue of the rat flaps in N-trimethyl chitosan-rTFPI group were significantly higher than those in N-trimethyl chitosan alone group (P<0.05 or P<0.01); the blood perfusion value in the middle segment tissue of the rat flaps in N-trimethyl chitosan-rTFPI group was significantly higher than that in rTFPI alone group (P<0.01). On POD 7, inflammatory cells infiltrated more and cell edema was obvious in the middle segment tissue of the rat flaps in PBS group and N-trimethyl chitosan alone group. Compared with those of the previous two groups, the inflammation degrees in the middle segment tissue of the rat flaps in rTFPI alone group and N-trimethyl chitosan-rTFPI group were significantly milder, the number of embolized blood vessels was significantly decreased (P<0.05 or P<0.01), and the number of new blood vessels was significantly increased (P<0.05 or P<0.01). Compared with that of rTFPI alone group, the number of new blood vessels in the middle segment tissue of the rat flaps in N-trimethyl chitosan-rTFPI group increased significantly (P<0.05). Conclusions: The effect of sustained release of rTFPI can be achieved by loading rTFPI with N-trimethyl chitosan. Compared with rTFPI alone, the N-trimethyl chitosan-rTFPI complex can further improve the blood perfusion of the avulsion flaps with roll compaction in rat and improve the survival rate of the flap.
Collapse
Affiliation(s)
- J F Wu
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| | - X D Hong
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| | - J Jin
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| | - Y H H Fei
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| | - M Y Zhang
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| | - T T Si
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| | - H Fan
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| | - X D Zhang
- Department of Burns and Plastic Surgery, the 903th Hospital of the Joint Logistics Support Force of the People's Liberation Army, Hangzhou 310013, China
| |
Collapse
|
162
|
Sheykhloo H, Milani M, Najafi F, Bani F, Zarebkohan A. Conjugation of Gentamicin to Polyamidoamine Dendrimers Improved Anti-bacterial Properties against Pseudomonas aeruginosa. Adv Pharm Bull 2021; 11:675-683. [PMID: 34888214 PMCID: PMC8642794 DOI: 10.34172/apb.2021.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/29/2020] [Accepted: 10/13/2020] [Indexed: 11/15/2022] Open
Abstract
Purpose: This study aimed to design gentamicin-conjugated poly (amidoamine) (PAMAM) dendrimers to increase the therapeutic efficiency of gentamicin against Pseudomonas aeruginosa. Methods: Gentamicin-presenting dendrimers were synthesized using MAL-PEG3400-NHS as a redox-sensitive linker to attach gentamicin to the surface of G4 PAMAM dendrimers. The gentamicin molecules were thiolated by using Traut reagent. Then, the functionalized gentamicin molecules were attached to PEGylated PAMAM dendrimers through simple and high selectively maleimide (MAL)-thiol reaction. The structure of gentamicin-conjugated PAMAM dendrimers was characterized and confirmed using nuclear magnetic resonance (NMR), dynamic light scattering (DLS), zeta potential analysis, and transmission electron microscopy (TEM) imaging. The antibacterial properties of the synthesized complex were examined on P. aeruginosa and compared to gentamycin alone. Results: NMR, DLS, zeta potential analysis, and TEM imaging revealed the successful conjugation of gentamicin to PAMAM dendrimers. Data showed the appropriate physicochemical properties of the synthesized nanoparticles. We found a 3-fold increase in the antibacterial properties of gentamicin conjugated to the surface of PAMAM dendrimers compared to non-conjugated gentamicin. Based on data, the anti-biofilm effects of PAMAM-Gentamicin dendrimers increased at least 13 times more than the gentamicin in normal conditions. Conclusion: Data confirmed that PAMAM dendrimer harboring gentamicin could be touted as a novel smart drug delivery system in infectious conditions.
Collapse
Affiliation(s)
- Hamed Sheykhloo
- Biotechnology Department, Rabe Rashidi University, Tabriz, Iran
| | - Morteza Milani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhood Najafi
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran, Iran
| | - Farhad Bani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
163
|
Luo G, Zhang J, Sun Y, Wang Y, Wang H, Cheng B, Shu Q, Fang X. Nanoplatforms for Sepsis Management: Rapid Detection/Warning, Pathogen Elimination and Restoring Immune Homeostasis. NANO-MICRO LETTERS 2021; 13:88. [PMID: 33717630 PMCID: PMC7938387 DOI: 10.1007/s40820-021-00598-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/14/2020] [Indexed: 05/20/2023]
Abstract
Sepsis, a highly life-threatening organ dysfunction caused by uncontrollable immune responses to infection, is a leading contributor to mortality in intensive care units. Sepsis-related deaths have been reported to account for 19.7% of all global deaths. However, no effective and specific therapeutic for clinical sepsis management is available due to the complex pathogenesis. Concurrently eliminating infections and restoring immune homeostasis are regarded as the core strategies to manage sepsis. Sophisticated nanoplatforms guided by supramolecular and medicinal chemistry, targeting infection and/or imbalanced immune responses, have emerged as potent tools to combat sepsis by supporting more accurate diagnosis and precision treatment. Nanoplatforms can overcome the barriers faced by clinical strategies, including delayed diagnosis, drug resistance and incapacity to manage immune disorders. Here, we present a comprehensive review highlighting the pathogenetic characteristics of sepsis and future therapeutic concepts, summarizing the progress of these well-designed nanoplatforms in sepsis management and discussing the ongoing challenges and perspectives regarding future potential therapies. Based on these state-of-the-art studies, this review will advance multidisciplinary collaboration and drive clinical translation to remedy sepsis.
Collapse
Affiliation(s)
- Gan Luo
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 People’s Republic of China
| | - Jue Zhang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 People’s Republic of China
| | - Yaqi Sun
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 People’s Republic of China
| | - Ya Wang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 People’s Republic of China
| | - Hanbin Wang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 People’s Republic of China
| | - Baoli Cheng
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 People’s Republic of China
| | - Qiang Shu
- National Clinical Research Center for Child Health, Children’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310052 People’s Republic of China
| | - Xiangming Fang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 People’s Republic of China
| |
Collapse
|
164
|
Mori T, Yoshida M, Hazekawa M, Ishibashi D, Hatanaka Y, Kakehashi R, Nakagawa M, Nagao T, Yoshii M, Kojima H, Uno R, Uchida T. Targeted Delivery of Miconazole Employing LL37 Fragment Mutant Peptide CKR12-Poly (Lactic-Co-Glycolic) Acid Polymeric Micelles. Int J Mol Sci 2021; 22:ijms222112056. [PMID: 34769486 PMCID: PMC8584378 DOI: 10.3390/ijms222112056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 11/17/2022] Open
Abstract
We previously reported that conjugates of antimicrobial peptide fragment analogues and poly (lactic-co-glycolic) acid (PLGA) enhance antimicrobial activity and that the conjugated micelle structure is an effective tool for antimicrobial drug delivery. In recent years, the delivery of antimicrobial peptides to targets for antimicrobial activity has attracted attention. In this study, we targeted Candida albicans, a causative organism of catheter-related bloodstream infections, which is refractory to antimicrobial agents and is currently a problem in medical practice. We evaluated the antifungal activity of CKR12 (a mutant fragment of the human cathelicidin peptide, LL-37)-PLGA-miconazole (MCZ) micelles using nanotechnology with MCZ delivery. The prepared CKR12-PLGA-MCZ micelles were characterised by measuring dynamic light scattering, zeta potential, dilution stability, and drug release. CKR12-PLGA-MCZ micelles showed higher antifungal activity than CKR12-PLGA micelles and MCZ solution. Furthermore, scanning and transmission electron microscopy suggested that CKR12-PLGA-MCZ micelles disrupted both cell wall and cell membrane of C. albicans. Our results revealed a synergistic effect of antifungal activity using a combination of antimicrobial peptide fragment analogues and MCZ, and that MCZ is a promising tool for the delivery to target microorganisms.
Collapse
Affiliation(s)
- Takeshi Mori
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Mukogawa Women’s University, 11-68 Koshien 9-Bancho, Nishinomiya City 663-8179, Hyogo, Japan; (T.M.); (M.Y.); (H.K.); (R.U.)
| | - Miyako Yoshida
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Mukogawa Women’s University, 11-68 Koshien 9-Bancho, Nishinomiya City 663-8179, Hyogo, Japan; (T.M.); (M.Y.); (H.K.); (R.U.)
| | - Mai Hazekawa
- Department of Immunological and Molecular Pharmacology, Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-Ku, Fukuoka City 814-0180, Fukuoka, Japan; (M.H.); (D.I.)
| | - Daisuke Ishibashi
- Department of Immunological and Molecular Pharmacology, Faculty of Pharmaceutical Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-Ku, Fukuoka City 814-0180, Fukuoka, Japan; (M.H.); (D.I.)
| | - Yoshiro Hatanaka
- Osaka Research Institute of Industrial Science and Technology, 1-6-50 Morinomiya, Joto-ku, Osaka City 536-8553, Osaka, Japan; (Y.H.); (R.K.); (M.N.); (T.N.); (M.Y.)
| | - Rie Kakehashi
- Osaka Research Institute of Industrial Science and Technology, 1-6-50 Morinomiya, Joto-ku, Osaka City 536-8553, Osaka, Japan; (Y.H.); (R.K.); (M.N.); (T.N.); (M.Y.)
| | - Makoto Nakagawa
- Osaka Research Institute of Industrial Science and Technology, 1-6-50 Morinomiya, Joto-ku, Osaka City 536-8553, Osaka, Japan; (Y.H.); (R.K.); (M.N.); (T.N.); (M.Y.)
| | - Toshihiro Nagao
- Osaka Research Institute of Industrial Science and Technology, 1-6-50 Morinomiya, Joto-ku, Osaka City 536-8553, Osaka, Japan; (Y.H.); (R.K.); (M.N.); (T.N.); (M.Y.)
| | - Miki Yoshii
- Osaka Research Institute of Industrial Science and Technology, 1-6-50 Morinomiya, Joto-ku, Osaka City 536-8553, Osaka, Japan; (Y.H.); (R.K.); (M.N.); (T.N.); (M.Y.)
| | - Honami Kojima
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Mukogawa Women’s University, 11-68 Koshien 9-Bancho, Nishinomiya City 663-8179, Hyogo, Japan; (T.M.); (M.Y.); (H.K.); (R.U.)
| | - Rio Uno
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Mukogawa Women’s University, 11-68 Koshien 9-Bancho, Nishinomiya City 663-8179, Hyogo, Japan; (T.M.); (M.Y.); (H.K.); (R.U.)
| | - Takahiro Uchida
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Mukogawa Women’s University, 11-68 Koshien 9-Bancho, Nishinomiya City 663-8179, Hyogo, Japan; (T.M.); (M.Y.); (H.K.); (R.U.)
- Correspondence: ; Tel.: +81-798-45-9957
| |
Collapse
|
165
|
Nwabuife JC, Pant AM, Govender T. Liposomal delivery systems and their applications against Staphylococcus aureus and Methicillin-resistant Staphylococcus aureus. Adv Drug Deliv Rev 2021; 178:113861. [PMID: 34242712 DOI: 10.1016/j.addr.2021.113861] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/16/2022]
Abstract
Liposomal delivery systems have been widely explored for targeting superbugs such as S. aureus and MRSA, overcoming antimicrobial resistance associated with conventional dosage forms. They have the significant advantage of delivering hydrophilic and lipophilic antimicrobial agents, either singularly as monotherapy or in combination as combination therapy, due to their bilayers with action-site-specificity, resulting in improved targeting compared to conventional dosage forms. Herein, we present an extensive and critical review of the different liposomal delivery systems employed in the past two decades for the delivery of both antibiotics of different classes and non-antibiotic antibacterial agents, as monotherapy and combination therapy to eradicate infections caused by S. aureus and MRSA. The review also identifies future research and strategies potentiating the applications of liposomal delivery systems against S. aureus and MRSA. This review confirms the potential application of liposomal delivery systems for effective delivery and specific targeting of S. aureus and MRSA infections.
Collapse
|
166
|
Muhammad W, Zhai Z, Wang S, Gao C. Inflammation-modulating nanoparticles for pneumonia therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1763. [PMID: 34713969 DOI: 10.1002/wnan.1763] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022]
Abstract
Pneumonia is a common but serious infectious disease, and is the sixth leading cause for death. The foreign pathogens such as viruses, fungi, and bacteria establish an inflammation response after interaction with lung, leading to the filling of bronchioles and alveoli with fluids. Although the pharmacotherapies have shown their great effectiveness to combat pathogens, advanced methods are under developing to treat complicated cases such as virus-infection and lung inflammation or acute lung injury (ALI). The inflammation modulation nanoparticles (NPs) can effectively suppress immune cells and inhibit inflammatory molecules in the lung site, and thereby alleviate pneumonia and ALI. In this review, the pathological inflammatory microenvironments in pneumonia, which are instructive for the design of biomaterials therapy, are summarized. The focus is then paid to the inflammation-modulating NPs that modulate the inflammatory cells, cytokines and chemokines, and microenvironments of pneumonia for better therapeutic effects. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Respiratory Disease.
Collapse
Affiliation(s)
- Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Shuqin Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
167
|
The Antibiofilm Nanosystems for Improved Infection Inhibition of Microbes in Skin. Molecules 2021; 26:molecules26216392. [PMID: 34770799 PMCID: PMC8587837 DOI: 10.3390/molecules26216392] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Biofilm formation is an important virulence factor for the opportunistic microorganisms that elicit skin infections. The recalcitrant feature of biofilms and their antibiotic tolerance impose a great challenge on the use of conventional therapies. Most antibacterial agents have difficulty penetrating the matrix produced by a biofilm. One novel approach to address these concerns is to prevent or inhibit the formation of biofilms using nanoparticles. The advantages of using nanosystems for antibiofilm applications include high drug loading efficiency, sustained or prolonged drug release, increased drug stability, improved bioavailability, close contact with bacteria, and enhanced accumulation or targeting to biomasses. Topically applied nanoparticles can act as a strategy for enhancing antibiotic delivery into the skin. Various types of nanoparticles, including metal oxide nanoparticles, polymeric nanoparticles, liposomes, and lipid-based nanoparticles, have been employed for topical delivery to treat biofilm infections on the skin. Moreover, nanoparticles can be designed to combine with external stimuli to produce magnetic, photothermal, or photodynamic effects to ablate the biofilm matrix. This study focuses on advanced antibiofilm approaches based on nanomedicine for treating skin infections. We provide in-depth descriptions on how the nanoparticles could effectively eliminate biofilms and any pathogens inside them. We then describe cases of using nanoparticles for antibiofilm treatment of the skin. Most of the studies included in this review were supported by in vivo animal infection models. This article offers an overview of the benefits of nanosystems for treating biofilms grown on the skin.
Collapse
|
168
|
Liu H, Zhong W, Zhang X, Lin D, Wu J. Nanomedicine as a promising strategy for the theranostics of infectious diseases. J Mater Chem B 2021; 9:7878-7908. [PMID: 34611689 DOI: 10.1039/d1tb01316e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Infectious diseases caused by bacteria, viruses, and fungi and their global spread pose a great threat to human health. The 2019 World Health Organization report predicted that infection-related mortality will be similar to cancer mortality by 2050. Particularly, the global cumulative numbers of the recent outbreak of coronavirus disease (COVID-19) have reached 110.7 million cases and over 2.4 million deaths as of February 23, 2021. Moreover, the crisis of these infectious diseases exposes the many problems of traditional diagnosis, treatment, and prevention, such as time-consuming and unselective detection methods, the emergence of drug-resistant bacteria, serious side effects, and poor drug delivery. There is an urgent need for rapid and sensitive diagnosis as well as high efficacy and low toxicity treatments. The emergence of nanomedicine has provided a promising strategy to greatly enhance detection methods and drug treatment efficacy. Owing to their unique optical, magnetic, and electrical properties, nanoparticles (NPs) have great potential for the fast and selective detection of bacteria, viruses, and fungi. NPs exhibit remarkable antibacterial activity by releasing reactive oxygen species and metal ions, exerting photothermal effects, and causing destruction of the cell membrane. Nano-based delivery systems can further improve drug permeability, reduce the side effects of drugs, and prolong systemic circulation time and drug half-life. Moreover, effective drugs against COVID-19 are still lacking. Recently, nanomedicine has shown great potential to accelerate the development of safe and novel anti-COVID-19 drugs. This article reviews the fundamental mechanisms and the latest developments in the treatment and diagnosis of bacteria, viruses, and fungi and discusses the challenges and perspectives in the application of nanomedicine.
Collapse
Affiliation(s)
- Hengyu Liu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Wenhao Zhong
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Xinyu Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jun Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China. .,School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
169
|
Kuzma BA, Pence IJ, Greenfield DA, Ho A, Evans CL. Visualizing and quantifying antimicrobial drug distribution in tissue. Adv Drug Deliv Rev 2021; 177:113942. [PMID: 34437983 DOI: 10.1016/j.addr.2021.113942] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022]
Abstract
The biodistribution and pharmacokinetics of drugs are vital to the mechanistic understanding of their efficacy. Measuring antimicrobial drug efficacy has been challenging as plasma drug concentration is used as a surrogate for tissue drug concentration, yet typically does not reflect that at the intended site(s) of action. Utilizing an image-guided approach, it is feasible to accurately quantify the biodistribution and pharmacokinetics within the desired site(s) of action. We outline imaging modalities used in visualizing drug distribution with examples ranging from in vitro cellular drug uptake to clinical treatment of microbial infections. The imaging modalities of interest are: radio-labeling, magnetic resonance, mass spectrometry imaging, computed tomography, fluorescence, and Raman spectroscopy. We outline the progress, limitations, and future outlook for each methodology. Further advances in these optical approaches would benefit patients and researchers alike, as non-invasive imaging could yield more profound insights with a lower clinical burden than invasive measurement approaches used today.
Collapse
Affiliation(s)
- Benjamin A Kuzma
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Isaac J Pence
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Daniel A Greenfield
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Alexander Ho
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Conor L Evans
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA.
| |
Collapse
|
170
|
Development of Chitosan/Cyclodextrin Nanospheres for Levofloxacin Ocular Delivery. Pharmaceutics 2021; 13:pharmaceutics13081293. [PMID: 34452254 PMCID: PMC8400911 DOI: 10.3390/pharmaceutics13081293] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/10/2021] [Accepted: 08/14/2021] [Indexed: 02/06/2023] Open
Abstract
Levofloxacin (LVF) is an antibacterial drug approved for the treatment of ocular infections. However, due to the low ocular bioavailability, high doses are needed, causing bacterial resistance. Polymeric nanospheres (NPs) loading antibiotic drugs represent the most promising approach to eradicate ocular infections and to treat pathogen resistance. In this study, we have developed chitosan NPs based on sulfobutyl-ether-β-cyclodextrin (CH/SBE-β-CD NPs) for ocular delivery of LVF. CH/SBE-β-CD NPs loading LVF were characterized in terms of encapsulation parameters, morphology, and sizes, in comparison to NPs produced without the macrocycle. Nuclear magnetic resonance and UV–vis spectroscopy studies demonstrated that SBE-β-CD is able to complex LVF and to influence encapsulation parameters of NPs, producing high encapsulation efficiency and LVF loading. The NPs were homogenous in size, with a hydrodynamic radius between 80 and 170 nm and positive zeta potential (ζ) values. This surface property could promote the interaction of NPs with the negatively charged ocular tissue, increasing their residence time and, consequently, LVF efficacy. In vitro, antibacterial activity against Gram-positive and Gram-negative bacteria showed a double higher activity of CH/SBE-β-CD NPs loading LVF compared to the free drug, suggesting that chitosan NPs based on SBE-β-CD could be a useful system for the treatment of ocular infections.
Collapse
|
171
|
Kanwal A, Uzair B, Sajjad S, Samin G, Ali Khan B, Khan Leghari SA, Khan Niazi MB, Abbas S. Synthesis and Characterization of Carbon Dots Coated CaCO 3 Nanocarrier for Levofloxacin Against Multidrug Resistance Extended-Spectrum Beta-Lactamase Escherichia coli of Urinary Tract Infection Origin. Microb Drug Resist 2021; 28:106-119. [PMID: 34402682 DOI: 10.1089/mdr.2020.0621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The multidrug resistance (MDR) Escherichia coli having Extended-Spectrum Beta-Lactamase (ESBL) genes and the capacity to create a biofilm acts as a major reduction in the therapeutic effectiveness of antimicrobials. In search of a novel nanocarrier (NC) for targeted delivery of antibiotics, carbon dots (CDs) coated calcium carbonate nanocarriers (CCNC) from organic chicken eggshells conjugated with levofloxacin (Lvx) were synthesized. Our main objectives were to explore the antimicrobial, antibiofilm, and NC potential of CDs coated CaCO3 Nanocarrier conjugated with levofloxacin (CD-CCNC-Lvx) to combat biofilm-producing MDR ESBL E. coli of urinary tract infection origin. The synthesized NC system was physiochemically characterized, validating the synthesis of CCNC and CD-CCNC-Lvx with a particle size of 56 and 14 nm, respectively. Scanning electron microscopy (SEM) showed rod shape morphology. X-ray diffraction results discovered crystalline and dispersed nanoparticles. In vitro release drug kinetics illustrated sustained release of Lvx. NC system exhibited strong antibacterial and antibiofilm potential against E. coli with a noticeable low minimal inhibitory concentration (MIC). MIC of CCNC was found to be 30 ± 0.1 μg/mL and CD-CCNC-Lvx was 20 ± 0.1 μg/mL for MDR ESBL-producing E. coli. The synergistic effect of NC upon conjugation with Lvx showed incredible activity with 30 mm zone of inhibition and 68% biofilm inhibition. Flow cytometry analysis revealed treated E. coli cells showed 58.69% reduction in cell viability. SEM images of treated bacterial cells showed morphological changes, which were also confirmed by our flow cytometry findings leading to cell membrane damage in E. coli. NC system also downregulated the blaCTX-M gene in E. coli. The hemolytic analysis proved biocompatibility with human red blood cells (RBCs). It is concluded that CCNC has the potential to be used as NC for target delivery of antibiotics and may combat toxicity of antibiotics as the inhibition of E. coli was noticed at low MIC concentration.
Collapse
Affiliation(s)
- Amna Kanwal
- Department of Biological Sciences and International Islamic University, Islamabad, Pakistan
| | - Bushra Uzair
- Department of Biological Sciences and International Islamic University, Islamabad, Pakistan
| | - Shamaila Sajjad
- Department of Physics, International Islamic University, Islamabad, Pakistan
| | - Ghufrana Samin
- Department of Chemistry, University of Engineering and Technology Lahore, Faisalabad Campus, Faisalabad, Pakistan
| | - Barkat Ali Khan
- Department of Pharmaceutics, Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | | | - Muhammad Bilal Khan Niazi
- School of Chemical & Materials Engineering, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sehrish Abbas
- Department of Biological Sciences and International Islamic University, Islamabad, Pakistan
| |
Collapse
|
172
|
Hu XL, Shang Y, Yan KC, Sedgwick AC, Gan HQ, Chen GR, He XP, James TD, Chen D. Low-dimensional nanomaterials for antibacterial applications. J Mater Chem B 2021; 9:3640-3661. [PMID: 33870985 DOI: 10.1039/d1tb00033k] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The excessive use of antibiotics has led to a rise in drug-resistant bacteria. These "superbugs" are continuously emerging and becoming increasingly harder to treat. As a result, new and effective treatment protocols that have minimal risks of generating drug-resistant bacteria are urgently required. Advanced nanomaterials are particularly promising due to their drug loading/releasing capabilities combined with their potential photodynamic/photothermal therapeutic properties. In this review, 0-dimensional, 1-dimensional, 2-dimensional, and 3-dimensional nanomaterial-based systems are comprehensively discussed for bacterial-based diagnostic and treatment applications. Since the use of these platforms as antibacterials is relatively new, this review will provide appropriate insight into their construction and applications. As such, we hope this review will inspire researchers to explore antibacterial-based nanomaterials with the aim of developing systems for clinical applications.
Collapse
Affiliation(s)
- Xi-Le Hu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Ying Shang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Kai-Cheng Yan
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | - Adam C Sedgwick
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, USA
| | - Hui-Qi Gan
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Guo-Rong Chen
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Rd, Shanghai 200237, China.
| | - Tony D James
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK. and School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China.
| |
Collapse
|
173
|
Bhattacharjee B, Ghosh S, Patra D, Haldar J. Advancements in release-active antimicrobial biomaterials: A journey from release to relief. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1745. [PMID: 34374498 DOI: 10.1002/wnan.1745] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/13/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022]
Abstract
Escalating medical expenses due to infectious diseases are causing huge socioeconomic pressure on mankind globally. The emergence of antibiotic resistance has further aggravated this problem. Drug-resistant pathogens are also capable of forming thick biofilms on biotic and abiotic surfaces to thrive in a harsh environment. To address these clinical problems, various strategies including antibacterial agent delivering matrices and bactericidal coatings strategies have been developed. In this review, we have discussed various types of polymeric vehicles such as hydrogels, sponges/cryogels, microgels, nanogels, and meshes, which are commonly used to deliver antibiotics, metal nanoparticles, and biocides. Compositions of these polymeric matrices have been elaborately depicted by elucidating their chemical interactions and potential activity have been discussed. On the other hand, various implant/device-surface coating strategies which exploit the release-active mechanism of bacterial killing are discussed in elaboration. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Brinta Bhattacharjee
- Antimicrobial Research Laboratory, New Chemistry, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru, Karnataka, India
| | - Sreyan Ghosh
- Antimicrobial Research Laboratory, New Chemistry, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru, Karnataka, India
| | - Dipanjana Patra
- Antimicrobial Research Laboratory, New Chemistry, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru, Karnataka, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru, Karnataka, India.,School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru, Karnataka, India
| |
Collapse
|
174
|
Schneider-Futschik EK, Reyes-Ortega F. Advantages and Disadvantages of Using Magnetic Nanoparticles for the Treatment of Complicated Ocular Disorders. Pharmaceutics 2021; 13:1157. [PMID: 34452117 PMCID: PMC8400382 DOI: 10.3390/pharmaceutics13081157] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 01/01/2023] Open
Abstract
Nanomaterials provide enormous opportunities to overcome the limitations of conventional ocular delivery systems, such as low therapeutic efficacy, side effects due to the systemic exposure, or invasive surgery. Apart from the more common ocular disorders, there are some genetic diseases, such as cystic fibrosis, that develop ocular disorders as secondary effects as long as the disease progresses. These patients are more difficult to be pharmacologically treated using conventional drug routes (topically, systemic), since specific pharmacological formulations can be incompatible, display increased toxicity, or their therapeutic efficacy decreases with the administration of different kind of chemical molecules. Magnetic nanoparticles can be used as potent drug carriers and magnetic hyperthermia agents due to their response to an external magnetic field. Drugs can be concentrated in the target point, limiting the damage to other tissues. The other advantage of these magnetic nanoparticles is that they can act as magnetic resonance imaging agents, allowing the detection of the exact location of the disease. However, there are some drawbacks related to their use in drug delivery, such as the limitation to maintain efficacy in the target organ once the magnetic field is removed from outside. Another disadvantage is the difficulty in maintaining the therapeutic action in three dimensions inside the human body. This review summarizes all the application possibilities related to magnetic nanoparticles in ocular diseases.
Collapse
Affiliation(s)
- Elena K. Schneider-Futschik
- Department of Biochemistry & Pharmacology, Faculty of Medicine, School of Biomedical Sciences, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Felisa Reyes-Ortega
- Visual Quality Research Group, Department of Ophthalmology, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital and University of Cordoba, 14004 Cordoba, Spain
| |
Collapse
|
175
|
Rinaldi F, Hanieh PN, Sennato S, De Santis F, Forte J, Fraziano M, Casciardi S, Marianecci C, Bordi F, Carafa M. Rifampicin-Liposomes for Mycobacterium abscessus Infection Treatment: Intracellular Uptake and Antibacterial Activity Evaluation. Pharmaceutics 2021; 13:1070. [PMID: 34371761 PMCID: PMC8309174 DOI: 10.3390/pharmaceutics13071070] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
Treatment of pulmonary infections caused by Mycobacterium abscessus are extremely difficult to treat, as this species is naturally resistant to many common antibiotics. Liposomes are vesicular nanocarriers suitable for hydrophilic and lipophilic drug loading, able to deliver drugs to the target site, and successfully used in different pharmaceutical applications. Moreover, liposomes are biocompatible, biodegradable and nontoxic vesicles and nebulized liposomes are efficient in targeting antibacterial agents to macrophages. The present aim was to formulate rifampicin-loaded liposomes (RIF-Lipo) for lung delivery, in order to increase the local concentration of the antibiotic. Unilamellar liposomal vesicles composed of anionic DPPG mixed with HSPC for rifampicin delivery were designed, prepared, and characterized. Samples were prepared by using the thin-film hydration method. RIF-Lipo and unloaded liposomes were characterized in terms of size, ζ-potential, bilayer features, stability and in different biological media. Rifampicin's entrapment efficiency and release were also evaluated. Finally, biological activity of RIF-loaded liposomes in Mycobacterium abscessus-infected macrophages was investigated. The results show that RIF-lipo induce a significantly better reduction of intracellular Mycobacterium abscessus viability than the treatment with free drug. Liposome formulation of rifampicin may represent a valuable strategy to enhance the biological activity of the drug against intracellular mycobacteria.
Collapse
Affiliation(s)
- Federica Rinaldi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma-Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.R.); (P.N.H.); (J.F.); (M.C.)
| | - Patrizia Nadia Hanieh
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma-Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.R.); (P.N.H.); (J.F.); (M.C.)
| | - Simona Sennato
- Istituto dei Sistemi Complessi (ISC)-CNR, sede “Sapienza” and Dipartimento di Fisica, Sapienza Università di Roma, 00185 Rome, Italy;
| | - Federica De Santis
- Dipartimento di Biologia, Università di Roma “Tor Vergata” Via della Ricerca Scientifica, 00133 Rome, Italy; (F.D.S.); (M.F.)
| | - Jacopo Forte
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma-Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.R.); (P.N.H.); (J.F.); (M.C.)
| | - Maurizio Fraziano
- Dipartimento di Biologia, Università di Roma “Tor Vergata” Via della Ricerca Scientifica, 00133 Rome, Italy; (F.D.S.); (M.F.)
| | - Stefano Casciardi
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, National Institute for Insurance against Accidents at Work (INAIL), Monteporzio Catone, 00144 Rome, Italy;
| | - Carlotta Marianecci
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma-Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.R.); (P.N.H.); (J.F.); (M.C.)
| | - Federico Bordi
- Istituto dei Sistemi Complessi (ISC)-CNR, sede “Sapienza” and Dipartimento di Fisica, Sapienza Università di Roma, 00185 Rome, Italy;
| | - Maria Carafa
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma-Piazzale Aldo Moro 5, 00185 Rome, Italy; (F.R.); (P.N.H.); (J.F.); (M.C.)
| |
Collapse
|
176
|
Yang X, Ye W, Qi Y, Ying Y, Xia Z. Overcoming Multidrug Resistance in Bacteria Through Antibiotics Delivery in Surface-Engineered Nano-Cargos: Recent Developments for Future Nano-Antibiotics. Front Bioeng Biotechnol 2021; 9:696514. [PMID: 34307323 PMCID: PMC8297506 DOI: 10.3389/fbioe.2021.696514] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
In the recent few decades, the increase in multidrug-resistant (MDR) bacteria has reached an alarming rate and caused serious health problems. The incidence of infections due to MDR bacteria has been accompanied by morbidity and mortality; therefore, tackling bacterial resistance has become an urgent and unmet challenge to be properly addressed. The field of nanomedicine has the potential to design and develop efficient antimicrobials for MDR bacteria using its innovative and alternative approaches. The uniquely constructed nano-sized antimicrobials have a predominance over traditional antibiotics because their small size helps them in better interaction with bacterial cells. Moreover, surface engineering of nanocarriers offers significant advantages of targeting and modulating various resistance mechanisms, thus owe superior qualities for overcoming bacterial resistance. This review covers different mechanisms of antibiotic resistance, application of nanocarrier systems in drug delivery, functionalization of nanocarriers, application of functionalized nanocarriers for overcoming bacterial resistance, possible limitations of nanocarrier-based approach for antibacterial delivery, and future of surface-functionalized antimicrobial delivery systems.
Collapse
Affiliation(s)
- Xinfu Yang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Wenxin Ye
- Department of Urology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yajun Qi
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhongni Xia
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
177
|
Skóra B, Krajewska U, Nowak A, Dziedzic A, Barylyak A, Kus-Liśkiewicz M. Noncytotoxic silver nanoparticles as a new antimicrobial strategy. Sci Rep 2021; 11:13451. [PMID: 34188097 PMCID: PMC8242066 DOI: 10.1038/s41598-021-92812-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/16/2021] [Indexed: 02/05/2023] Open
Abstract
Drug-resistance of bacteria is an ongoing problem in hospital treatment. The main mechanism of bacterial virulency in human infections is based on their adhesion ability and biofilm formation. Many approaches have been invented to overcome this problem, i.e. treatment with antibacterial biomolecules, which have some limitations e.g. enzymatic degradation and short shelf stability. Silver nanoparticles (AgNPs) may be alternative to these strategies due to their unique and high antibacterial properties. Herein, we report on yeast Saccharomyces cerevisiae extracellular-based synthesis of AgNPs. Transmission electron microscopy (TEM) revealed the morphology and structure of the metallic nanoparticles, which showed a uniform distribution and good colloid stability, measured by hydrodynamic light scattering (DLS). The energy dispersive X-ray spectroscopy (EDS) of NPs confirms the presence of silver and showed that sulfur-rich compounds act as a capping agent being adsorbed on the surface of AgNPs. Antimicrobial tests showed that AgNPs inhibit the bacteria growth, while have no impact on fungi growth. Moreover, tested NPs was characterized by high inhibitory potential of bacteria biofilm formation but also eradication of established biofilms. The cytotoxic effect of the NPs on four mammalian normal and cancer cell lines was tested through the metabolic activity, cell viability and wound-healing assays. Last, but not least, ability to deep penetration of the silver colloid to the root canal was imaged by scanning electron microscopy (SEM) to show its potential as the material for root-end filling.
Collapse
Affiliation(s)
- Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225, Rzeszów, Poland
| | - Urszula Krajewska
- Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszów, Poland
| | - Anna Nowak
- Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszów, Poland
| | - Andrzej Dziedzic
- College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszow, Poland
| | - Adriana Barylyak
- Laser Department Center of Imlantation and Prosthetic Dentistry "MM", Department of Therapeutical Dentistry, Lviv National Medical University Ukraine, Lviv, Poland
| | - Małgorzata Kus-Liśkiewicz
- Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszów, Poland.
| |
Collapse
|
178
|
Patel A, Dey S, Shokeen K, Karpiński TM, Sivaprakasam S, Kumar S, Manna D. Sulfonium-based liposome-encapsulated antibiotics deliver a synergistic antibacterial activity. RSC Med Chem 2021; 12:1005-1015. [PMID: 34223166 PMCID: PMC8221259 DOI: 10.1039/d1md00091h] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/01/2021] [Indexed: 12/29/2022] Open
Abstract
The devastating antibacterial infections, coupled with their antibiotic resistance abilities, emphasize the need for effective antibacterial therapeutics. In this prospect, liposomal delivery systems have been employed in improving the efficacy of the antibacterial agents. The liposome-based antibiotics enhance the therapeutic potential of the new or existing antibiotics and reduce their adverse effects. The current study describes the development of sulfonium-based antibacterial lipids that demonstrate the delivery of existing antibiotics. The presence of cationic sulfonium moieties and inherent membrane targeting abilities of the lipids could help reduce the antibiotic resistance abilities of the bacteria and deliver the antibiotics to remove the infectious pathogens electively. The transmission electron microscopic images and dynamic light scattering analyses revealed the liposome formation abilities of the sulfonium-based amphiphilic compounds in the aqueous medium. The effectiveness of the compounds was tested against the Gram-negative and Gram-positive bacterial strains. The viability of the bacterial cells was remarkably reduced in the presence of the compounds. The sulfonium-based compounds with pyridinium moiety and long hydrocarbon chains showed the most potent antibacterial activities among the tested compounds. Mechanistic studies revealed the membrane-targeted bactericidal activities of the compounds. The potent compound also showed tetracycline and amoxicillin encapsulation and sustained release profiles in the physiologically relevant medium. The tetracycline and amoxicillin-encapsulated lipid showed much higher antibacterial activities than the free antibiotics at similar concentrations, emphasizing the usefulness of the synergistic effect of sulfonium-based lipid and the antibiotics, signifying that the sulfonium lipid penetrated the bacterial membrane and increased the cellular uptake of the antibiotics. The potent lipid also showed therapeutic potential, as it is less toxic to mammalian cells (like HeLa and HaCaT cells) at concentrations higher than their minimum inhibitory concentration values against S. aureus, E. coli, and MRSA. Hence, the sulfonium-based lipid exemplifies a promising framework for assimilating various warheads, and provides a potent antibacterial material.
Collapse
Affiliation(s)
- Anjali Patel
- Indian Institute of Technology Guwahati, Centre for the Environment Guwahati Assam India
| | - Subhasis Dey
- Biological Chemistry Laboratory, Indian Institute of Technology Guwahati, Chemistry Guwahati Assam India
| | - Kamal Shokeen
- Indian Institute of Technology Guwahati, Biosciences and Bioengineering Guwahati Assam India
| | - Tomasz M Karpiński
- Department of Medical Microbiology, Poznań University of Medical Sciences Wieniawskiego Poznań Poland
| | | | - Sachin Kumar
- Indian Institute of Technology Guwahati, Biosciences and Bioengineering Guwahati Assam India
| | - Debasis Manna
- Indian Institute of Technology Guwahati, Centre for the Environment Guwahati Assam India
- Biological Chemistry Laboratory, Indian Institute of Technology Guwahati, Chemistry Guwahati Assam India
| |
Collapse
|
179
|
Owais HM, Baddour MM, El-Metwally HAER, Barakat HS, Ammar NS, Meheissen MA. Assessment of the in vitro activity of azithromycin niosomes alone and in combination with levofloxacin on extensively drug-resistant Klebsiella pneumoniae clinical isolates. Braz J Microbiol 2021; 52:597-606. [PMID: 33483896 PMCID: PMC8105464 DOI: 10.1007/s42770-021-00433-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/11/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND AIM Extensively drug-resistant (XDR) Klebsiella pneumoniae represent a major threat in intensive care units. The aim of the current study was to formulate a niosomal form of azithromycin (AZM) and to evaluate its in vitro effect on XDR K. pneumoniae as a single agent or in combination with levofloxacin. MATERIAL AND METHODS Forty XDR K. pneumoniae isolates (23 colistin-sensitive and 17 colistin-resistant) were included in the study. Formulation and characterization of AZM niosomes were performed. The in vitro effect of AZM solution/niosomes alone and in combination (with levofloxacin) was investigated using the checkerboard assay, confirmed with time-kill assay and post-antibiotic effect (PAE). RESULTS The AZM niosome mean minimal inhibitory concentration (MIC) (187.4 ± 209.1 μg/mL) was significantly lower than that of the AZM solution (342.5 ± 343.4 μg/mL). AZM niosomes/levofloxacin revealed a 40% synergistic effect compared to 20% with AZM solution/levofloxacin. No antagonistic effect was detected. The mean MIC values of both AZM niosomes and AZM solution were lower in the colistin-resistant group than in the colistin-sensitive group. The mean PAE time of AZM niosomes (2.3 ± 1.09 h) was statistically significantly longer than that of the AZM solution (1.37 ± 0.5 h) (p = 0.023). CONCLUSION AZM niosomes were proved to be more effective than AZM solution against XDR K. pneumoniae, even colistin-resistant isolates.
Collapse
Affiliation(s)
- Hoda Mohamed Owais
- Medical Microbiology and Immunology Department, Faculty of Medicine, Alexandria University, Khartoum Square, Azarita, Alexandria, 21512, Egypt
| | - Manal Mohammad Baddour
- Medical Microbiology and Immunology Department, Faculty of Medicine, Alexandria University, Khartoum Square, Azarita, Alexandria, 21512, Egypt
| | - Hala Abd El-Raouf El-Metwally
- Medical Microbiology and Immunology Department, Faculty of Medicine, Alexandria University, Khartoum Square, Azarita, Alexandria, 21512, Egypt
| | - Heba Soliman Barakat
- Pharmaceutics Department, Faculty of Pharmacy, Alexandria University, Alexandria, 21512, Egypt
| | - Nour Sherif Ammar
- Alexandria University Hospitals, Alexandria University, Alexandria, 21512, Egypt
| | - Marwa Ahmed Meheissen
- Medical Microbiology and Immunology Department, Faculty of Medicine, Alexandria University, Khartoum Square, Azarita, Alexandria, 21512, Egypt.
| |
Collapse
|
180
|
Antibiofilm and antivirulence potential of silver nanoparticles against multidrug-resistant Acinetobacter baumannii. Sci Rep 2021; 11:10751. [PMID: 34031472 PMCID: PMC8144575 DOI: 10.1038/s41598-021-90208-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/26/2021] [Indexed: 11/08/2022] Open
Abstract
We aimed to isolate Acinetobacter baumannii (A. baumannii) from wound infections, determine their resistance and virulence profile, and assess the impact of Silver nanoparticles (AgNPs) on the bacterial growth, virulence and biofilm-related gene expression. AgNPs were synthesized and characterized using TEM, XRD and FTIR spectroscopy. A. baumannii (n = 200) were isolated and identified. Resistance pattern was determined and virulence genes (afa/draBC, cnf1, cnf2, csgA, cvaC, fimH, fyuA, ibeA, iutA, kpsMT II, PAI, papC, PapG II, III, sfa/focDE and traT) were screened using PCR. Biofilm formation was evaluated using Microtiter plate method. Then, the antimicrobial activity of AgNPs was evaluated by the well-diffusion method, growth kinetics and MIC determination. Inhibition of biofilm formation and the ability to disperse biofilms in exposure to AgNPs were evaluated. The effect of AgNPs on the expression of virulence and biofilm-related genes (bap, OmpA, abaI, csuA/B, A1S_2091, A1S_1510, A1S_0690, A1S_0114) were estimated using QRT-PCR. In vitro infection model for analyzing the antibacterial activity of AgNPs was done using a co-culture infection model of A. baumannii with human fibroblast skin cell line HFF-1 or Vero cell lines. A. baumannii had high level of resistance to antibiotics. Most of the isolates harbored the fimH, afa/draBC, cnf1, csgA and cnf2, and the majority of A. baumannii produced strong biofilms. AgNPs inhibited the growth of A. baumannii efficiently with MIC ranging from 4 to 25 µg/ml. A. baumannii showed a reduced growth rate in the presence of AgNPs. The inhibitory activity and the anti-biofilm activity of AgNPs were more pronounced against the weak biofilm producers. Moreover, AgNPs decreased the expression of kpsMII , afa/draBC,bap, OmpA, and csuA/B genes. The in vitro infection model revealed a significant antibacterial activity of AgNPs against extracellular and intracellular A. baumannii. AgNPs highly interrupted bacterial multiplication and biofilm formation. AgNPs downregulated the transcription level of important virulence and biofilm-related genes. Our findings provide an additional step towards understanding the mechanisms by which sliver nanoparticles interfere with the microbial spread and persistence.
Collapse
|
181
|
Khan F, Bamunuarachchi NI, Pham DTN, Tabassum N, Khan MSA, Kim YM. Mixed biofilms of pathogenic Candida-bacteria: regulation mechanisms and treatment strategies. Crit Rev Microbiol 2021; 47:699-727. [PMID: 34003065 DOI: 10.1080/1040841x.2021.1921696] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mixed-species biofilm is one of the most frequently recorded clinical problems. Mixed biofilms develop as a result of interactions between microorganisms of a single or multiple species (e.g. bacteria and fungi). Candida spp., particularly Candida albicans, are known to associate with various bacterial species to form a multi-species biofilm. Mixed biofilms of Candida spp. have been previously detected in vivo and on the surfaces of many biomedical instruments. Treating infectious diseases caused by mixed biofilms of Candida and bacterial species has been challenging due to their increased resistance to antimicrobial drugs. Here, we review and discuss the clinical significance of mixed Candida-bacteria biofilms as well as the signalling mechanisms involved in Candida-bacteria interactions. We also describe possible approaches for combating infections associated with mixed biofilms, such as the use of natural or synthetic drugs and combination therapy. The review presented here is expected to contribute to the advances in the biomedical field on the understanding of underlying interaction mechanisms of pathogens in mixed biofilm, and alternative approaches to treating the related infections.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea
| | - Nilushi Indika Bamunuarachchi
- Department of Food Science and Technology, Pukyong National University, Busan, South Korea.,Department of Fisheries and Marine Sciences, Ocean University of Sri Lanka, Tangalle, Sri Lanka
| | - Dung Thuy Nguyen Pham
- Center of Excellence for Biochemistry and Natural Products, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam.,NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan, South Korea
| | - Mohd Sajjad Ahmad Khan
- Department of Basic Sciences, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea.,Department of Food Science and Technology, Pukyong National University, Busan, South Korea
| |
Collapse
|
182
|
Jiang L, Loo SCJ. Intelligent Nanoparticle-Based Dressings for Bacterial Wound Infections. ACS APPLIED BIO MATERIALS 2021; 4:3849-3862. [PMID: 34056562 PMCID: PMC8155196 DOI: 10.1021/acsabm.0c01168] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022]
Abstract
Conventional wound dressing materials containing free antibiotics for bacterial wound infections are presented with several limitations, that is, lack of controlled and triggered release capabilities, and may often not be adequate to address the complex bacteria microenvironment of such infections. Additionally, the improper usage of antibiotics may also result in the emergence of drug resistant strains. While delivery systems (i.e., nanoparticles) that encapsulate antibiotics may potentially overcome some of these limitations, their therapeutic outcomes are still less than desirable. For example, premature drug release or unintended drug activation may occur, which would greatly reduce treatment efficacy. To address this, responsive nanoparticle-based antimicrobial therapies could be a promising strategy. Such nanoparticles can be functionalized to react to a single stimulus or multi stimulus within the bacteria microenvironment and subsequently elicit a therapeutic response. Such "intelligent" nanoparticles can be designed to respond to the microenvironment, that is, an acidic pH, the presence of specific enzymes, bacterial toxins, etc. or to an external stimulus, for example, light, thermal, etc. These responsive nanoparticles can be further incorporated into wound dressings to better promote wound healing. This review summarizes and highlights the recent progress on such intelligent nanoparticle-based dressings as potential wound dressings for bacteria-infected wounds, along with the current challenges and prospects for these technologies to be successfully translated into the clinic.
Collapse
Affiliation(s)
- Lai Jiang
- School
of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Say Chye Joachim Loo
- School
of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Harvard
T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
183
|
Nader D, Yousef F, Kavanagh N, Ryan BK, Kerrigan SW. Targeting Internalized Staphylococcus aureus Using Vancomycin-Loaded Nanoparticles to Treat Recurrent Bloodstream Infections. Antibiotics (Basel) 2021; 10:antibiotics10050581. [PMID: 34068975 PMCID: PMC8156000 DOI: 10.3390/antibiotics10050581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/29/2021] [Accepted: 05/11/2021] [Indexed: 12/29/2022] Open
Abstract
The bacterial pathogen Staphylococcus aureus is a leading cause of bloodstream infections, where patients often suffer from relapse despite antibiotic therapy. Traditional anti-staphylococcal drugs display reduced effectivity against internalised bacteria, but nanoparticles conjugated with antibiotics can overcome these challenges. In the present study, we aimed to characterise the internalisation and re-emergence of S. aureus from human endothelial cells and construct a new formulation of nanoparticles that target intracellular bacteria. Using an in vitro infection model, we demonstrated that S. aureus invades and persists within endothelial cells, mediated through bacterial extracellular surface adhesion, Fibronectin-binding protein A/B. After internalising, S. aureus localises to vacuoles as determined by transmission electron microscopy. Viable S. aureus emerges from endothelial cells after 48 h, supporting the notion that intracellular persistence contributes to infection relapses during bloodstream infections. Poly lactic-co-glycolic acid nanoparticles were formulated using a water-in-oil double emulsion method, which loaded 10% vancomycin HCl with 82.85% ± 12 encapsulation efficiency. These non-toxic nanoparticles were successfully taken up by cells and demonstrated a biphasic controlled release of 91 ± 4% vancomycin. They significantly reduced S. aureus intracellular growth within infected endothelial cells, which suggests future potential applications for targeting internalised bacteria and reducing mortality associated with bacteraemia.
Collapse
Affiliation(s)
- Danielle Nader
- Cardiovascular Infection Research Group, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephens Green, Dublin 2, Ireland; (F.Y.); (N.K.)
- Correspondence: (D.N.); (S.W.K.); Tel.: +353-1-402-2104 (S.W.K.)
| | - Fajer Yousef
- Cardiovascular Infection Research Group, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephens Green, Dublin 2, Ireland; (F.Y.); (N.K.)
| | - Nicola Kavanagh
- Cardiovascular Infection Research Group, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephens Green, Dublin 2, Ireland; (F.Y.); (N.K.)
| | - Benedict K. Ryan
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephens Green, Dublin 2, Ireland;
| | - Steven W. Kerrigan
- Cardiovascular Infection Research Group, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, 123 St. Stephens Green, Dublin 2, Ireland; (F.Y.); (N.K.)
- Correspondence: (D.N.); (S.W.K.); Tel.: +353-1-402-2104 (S.W.K.)
| |
Collapse
|
184
|
Mamun MM, Sorinolu AJ, Munir M, Vejerano EP. Nanoantibiotics: Functions and Properties at the Nanoscale to Combat Antibiotic Resistance. Front Chem 2021; 9:687660. [PMID: 34055750 PMCID: PMC8155581 DOI: 10.3389/fchem.2021.687660] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
One primary mechanism for bacteria developing resistance is frequent exposure to antibiotics. Nanoantibiotics (nAbts) is one of the strategies being explored to counteract the surge of antibiotic resistant bacteria. nAbts are antibiotic molecules encapsulated with engineered nanoparticles (NPs) or artificially synthesized pure antibiotics with a size range of ≤100 nm in at least one dimension. NPs may restore drug efficacy because of their nanoscale functionalities. As carriers and delivery agents, nAbts can reach target sites inside a bacterium by crossing the cell membrane, interfering with cellular components, and damaging metabolic machinery. Nanoscale systems deliver antibiotics at enormous particle number concentrations. The unique size-, shape-, and composition-related properties of nAbts pose multiple simultaneous assaults on bacteria. Resistance of bacteria toward diverse nanoscale conjugates is considerably slower because NPs generate non-biological adverse effects. NPs physically break down bacteria and interfere with critical molecules used in bacterial processes. Genetic mutations from abiotic assault exerted by nAbts are less probable. This paper discusses how to exploit the fundamental physical and chemical properties of NPs to restore the efficacy of conventional antibiotics. We first described the concept of nAbts and explained their importance. We then summarized the critical physicochemical properties of nAbts that can be utilized in manufacturing and designing various nAbts types. nAbts epitomize a potential Trojan horse strategy to circumvent antibiotic resistance mechanisms. The availability of diverse types and multiple targets of nAbts is increasing due to advances in nanotechnology. Studying nanoscale functions and properties may provide an understanding in preventing future outbreaks caused by antibiotic resistance and in developing successful nAbts.
Collapse
Affiliation(s)
- M. Mustafa Mamun
- Center for Environmental Nanoscience and Risk, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, United States
| | - Adeola Julian Sorinolu
- Civil and Environmental Engineering, The William States Lee College of Engineering, University of North Carolina, Charlotte, NC, United States
| | - Mariya Munir
- Civil and Environmental Engineering, The William States Lee College of Engineering, University of North Carolina, Charlotte, NC, United States
| | - Eric P. Vejerano
- Center for Environmental Nanoscience and Risk, Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
185
|
Pandit S, Rahimi S, Derouiche A, Boulaoued A, Mijakovic I. Sustained release of usnic acid from graphene coatings ensures long term antibiofilm protection. Sci Rep 2021; 11:9956. [PMID: 33976310 PMCID: PMC8113508 DOI: 10.1038/s41598-021-89452-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Protecting surfaces from bacterial colonization and biofilm development is an important challenge for the medical sector, particularly when it comes to biomedical devices and implants that spend longer periods in contact with the human body. A particularly difficult challenge is ensuring long-term protection, which is usually attempted by ensuring sustained release of antibacterial compounds loaded onto various coatings. Graphene have a considerable potential to reversibly interact water insoluble molecules, which makes them promising cargo systems for sustained release of such compounds. In this study, we developed graphene coatings that act as carriers capable of sustained release of usnic acid (UA), and hence enable long-term protection of surfaces against colonization by bacterial pathogens Staphylococcus aureus and Staphylococcus epidermidis. Our coatings exhibited several features that made them particularly effective for antibiofilm protection: (i) UA was successfully integrated with the graphene material, (ii) a steady release of UA was documented, (iii) steady UA release ensured strong inhibition of bacterial biofilm formation. Interestingly, even after the initial burst release of UA, the second phase of steady release was sufficient to block bacterial colonization. Based on these results, we propose that graphene coatings loaded with UA can serve as effective antibiofilm protection of biomedical surfaces.
Collapse
Affiliation(s)
- Santosh Pandit
- grid.5371.00000 0001 0775 6028Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 41296 Göteborg, Sweden
| | - Shadi Rahimi
- grid.5371.00000 0001 0775 6028Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 41296 Göteborg, Sweden
| | - Abderahmane Derouiche
- grid.5371.00000 0001 0775 6028Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 41296 Göteborg, Sweden
| | - Athmane Boulaoued
- grid.5371.00000 0001 0775 6028Department of Physics, Chalmers University of Technology, Kemivägen 10, 41296 Göteborg, Sweden
| | - Ivan Mijakovic
- grid.5371.00000 0001 0775 6028Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 41296 Göteborg, Sweden ,grid.5170.30000 0001 2181 8870Center for Biosustainability, Novo Nordisk Foundation, Technical University of Denmark, Kongens, Lyngby, Denmark
| |
Collapse
|
186
|
Deusenbery C, Wang Y, Shukla A. Recent Innovations in Bacterial Infection Detection and Treatment. ACS Infect Dis 2021; 7:695-720. [PMID: 33733747 DOI: 10.1021/acsinfecdis.0c00890] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bacterial infections are a major threat to human health, exacerbated by increasing antibiotic resistance. These infections can result in tremendous morbidity and mortality, emphasizing the need to identify and treat pathogenic bacteria quickly and effectively. Recent developments in detection methods have focused on electrochemical, optical, and mass-based biosensors. Advances in these systems include implementing multifunctional materials, microfluidic sampling, and portable data-processing to improve sensitivity, specificity, and ease of operation. Concurrently, advances in antibacterial treatment have largely focused on targeted and responsive delivery for both antibiotics and antibiotic alternatives. Antibiotic alternatives described here include repurposed drugs, antimicrobial peptides and polymers, nucleic acids, small molecules, living systems, and bacteriophages. Finally, closed-loop therapies are combining advances in the fields of both detection and treatment. This review provides a comprehensive summary of the current trends in detection and treatment systems for bacterial infections.
Collapse
Affiliation(s)
- Carly Deusenbery
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, Rhode Island 02912, United States
| | - Yingying Wang
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
187
|
Shaheen S, Maqbool K, Gul F, Sohail A. EFFECT OF CHEMICAL REACTION AND THERMAL RADIATION ON AXISYMMETRIC MHD FLOW OF JEFFREY NANOFLUID THROUGH A CILIATED CHANNEL FILLED WITH POROUS MEDIUM. BIOMEDICAL ENGINEERING: APPLICATIONS, BASIS AND COMMUNICATIONS 2021. [DOI: 10.4015/s1016237221500253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
To prevent the respiratory diseases in an air ways, a defense mechanism based on mucus transport by the moving cilia plays an important role. The mucus transport is affected by the thermal radiation, chemical reaction that changes the physics of fluid due to nanoparticles and thickness of mucus, also different problems in respiratory tract may occur due to the mucus efficacy. In this study, it is observed that the mucus transport can be controlled by the magnetic field that is produced by the drug delivery of nanoparticles, thermal radiation due to temperature difference, porous medium due to respiratory infection, and diffusion of the nanoparticles (chemical reaction) due to the magnetic drug delivery. In this model, flow of Jeffrey nanofluid through the ciliated tube resembles with the mucus flow in a wind pipe. The movement of the mucus is observed by the momentum, energy and concentration equation in the presence of body forces due to magnetic field, heat source due to radiation, Darcy’s resistance due to infection and chemical reaction due to the concentration of nanoparticles. Mathematical model of this study forms a complex system of partial differential equations under the low Reynolds number and long wavelength approximation. The nonlinear set of partial differential equations is solved by the Homotopy perturbation method and software “Mathematica,” results are found for velocity, temperature and concentration profiles and concluded that the mucus flow decelerates due to magnetic field produced by the drug delivery of the nanoparticles but accelerates due to the viscoelastic parameter of Jeffrey fluid and Darcy’s resistance parameter due to infection. The heat transfer rate in the mucus flow rises by increasing the random motion and reduces by the radiation and energy loss. The diffusion of the nanoparticles in the mucus rises by the growing values of thermophoresis and chemical reaction parameter and reduces by the growing values of viscoelastic and Brownian motion parameter.
Collapse
Affiliation(s)
- Sidra Shaheen
- Department of Mathematics & Statistics, International Islamic University, Islamabad 44000, Pakistan
| | - Khadija Maqbool
- Department of Mathematics & Statistics, International Islamic University, Islamabad 44000, Pakistan
| | - Farah Gul
- Department of Mathematics & Statistics, International Islamic University, Islamabad 44000, Pakistan
| | - Ayesha Sohail
- Department of Mathematics, COMSATS Institute of Information Technology, Lahore, Pakistan
| |
Collapse
|
188
|
Spirescu VA, Chircov C, Grumezescu AM, Andronescu E. Polymeric Nanoparticles for Antimicrobial Therapies: An Up-To-Date Overview. Polymers (Basel) 2021; 13:724. [PMID: 33673451 PMCID: PMC7956825 DOI: 10.3390/polym13050724] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the many advancements in the pharmaceutical and medical fields and the development of numerous antimicrobial drugs aimed to suppress and destroy pathogenic microorganisms, infectious diseases still represent a major health threat affecting millions of lives daily. In addition to the limitations of antimicrobial drugs associated with low transportation rate, water solubility, oral bioavailability and stability, inefficient drug targeting, considerable toxicity, and limited patient compliance, the major cause for their inefficiency is the antimicrobial resistance of microorganisms. In this context, the risk of a pre-antibiotic era is a real possibility. For this reason, the research focus has shifted toward the discovery and development of novel and alternative antimicrobial agents that could overcome the challenges associated with conventional drugs. Nanotechnology is a possible alternative, as there is significant evidence of the broad-spectrum antimicrobial activity of nanomaterials and nanoparticles in particular. Moreover, owing to their considerable advantages regarding their efficient cargo dissolving, entrapment, encapsulation, or surface attachment, the possibility of forming antimicrobial groups for specific targeting and destruction, biocompatibility and biodegradability, low toxicity, and synergistic therapy, polymeric nanoparticles have received considerable attention as potential antimicrobial drug delivery agents. In this context, the aim of this paper is to provide an up-to-date overview of the most recent studies investigating polymeric nanoparticles designed for antimicrobial therapies, describing both their targeting strategies and their effects.
Collapse
Affiliation(s)
- Vera Alexandra Spirescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Cristina Chircov
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania; (V.A.S.); (C.C.); (E.A.)
| |
Collapse
|
189
|
Wei X, Li P, Zhou H, Hu X, Liu D, Wu J, Wang Y. Engineering of gemcitabine coated nano-graphene oxide sheets for efficient near-infrared radiation mediated in vivo lung cancer photothermal therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 216:112125. [PMID: 33601257 DOI: 10.1016/j.jphotobiol.2021.112125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 01/02/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022]
Abstract
Gemcitabine (GEM) and its derivatives of deoxycytosine is a promising anticancer candidate which is effective for the treatment of various cancers including lung cancer via cascade targetting Erk/Mek/Raf/Ras pathway and blocking the proliferation of the tumor cells. In this present work, we have described reduced graphene oxide (rGO) in the presence of anticancer utilizing ascorbic acid as reducing agents for lung cancer treatment. GEM reduced graphene oxide (termed as GEM-rGO) has resulted in a smooth and transparent morphological surface, which was confirmed by various spectroscopical investigations. The anticancer drug-loaded rGO has displayed remarkable cytotoxic activities against a panel of lung cancer cell lines when compared to the untreated lung cancer cells. Further, we examined the morphological observation of the cancer cell death was monitored through the fluorescence microscopic examinations. In addition, the cell deaths of the lung cancer cells were observed by the flow cytometry analyses. In addition, the non-toxic nature of potent GEM-rGO and GEM-rGO + NIR was confirmed by in vivo systemic toxicity analysis. Besides, the higher safety feature of the GEM-rGO and GEM-rGO + NIR was evidenced by histological analyses of the mice organs. The subcutaneous injection of GEM-rGO and GEM-rGO + NIR into mice bearing A549 xenografts more effectively inhibited the tumor than the free GEM. Based on the outcomes, we can summarise that the GEM reduced graphene oxide (GEM-rGO) can be used as a promising drug candidate for the treatment of lung cancer in the future.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Peixian Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Hongfeng Zhou
- Department of Medical Oncology, General Hospital of Heilongjiang Province Land Reclamation Bureau, Harbin 150088, Heilongjiang, China
| | - Xiaowei Hu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Dan Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Jin Wu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Yi Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
190
|
Emerging trends in aggregation induced emissive luminogens as bacterial theranostics. J Drug Target 2021; 29:793-807. [PMID: 33583291 DOI: 10.1080/1061186x.2021.1888111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The emergence and spread of pathogenic bacteria, particularly antibiotic-resistant strains pose grave global concerns worldwide, which demand for the rapid development of highly selective and sensitive strategies for specific bacterial detection, identification, imaging and therapy. The fascinating feature of aggregation-induced emissive molecules (AIEgens) to display fluorescence in aggregate form can be suitably coupled with nanotechnology for developing theranostic AIE dots that can offer convenient and customised functions such as sensing, imaging, detection, discrimination and cell kill of different bacterial types. The initial section of the article reveals the necessity for incorporating diagnostic imaging with antibacterial therapy, while the latter part delivers mechanistic insights on the benefits of AIE fluorophores in theranostic applications. Further, the review illustrates the recent advancements of AIEgens as theranostic nanolights in bacterial detection, identification and eradication. The review is organised according to the different classes of AIE-active bacterial theranostics such as carrier-free nanoprodrugs, nanomachines for synergistic imaging-guided cancer treatment and bacterial kill, AIE polymers, bioconjugates and nanoparticle carriers. By elucidating their design principles and applications, as well as highlighting the recent trends and perspectives that can be further explored, we hope to instill more research interest in AIE bacterial theranostics for future translational research.HighlightsCombination of aggregation induced emissive fluorophores and nanotechnology for developing bacterial theranostics.AIE theranostics with customised functions for bacterial imaging, detection, discrimination and cell kill.
Collapse
|
191
|
Liao CC, Yu HP, Yang SC, Alalaiwe A, Dai YS, Liu FC, Fang JY. Multifunctional lipid-based nanocarriers with antibacterial and anti-inflammatory activities for treating MRSA bacteremia in mice. J Nanobiotechnology 2021; 19:48. [PMID: 33588861 PMCID: PMC7885212 DOI: 10.1186/s12951-021-00789-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/02/2021] [Indexed: 01/12/2023] Open
Abstract
Background Bacteremia-induced sepsis is a leading cause of mortality in intensive care units. To control a bacterial infection, an immune response is required, but this response might contribute to organ failure. Kidneys are one of the main organs affected by bacteremia. Combination therapies with antibacterial and anti-inflammatory effects may be beneficial in treating bacteremia. This study aimed to develop nanostructured lipid carriers (NLCs) loaded with ciprofloxacin and rolipram that exert a combination of anti-methicillin-resistant Staphylococcus aureus (MRSA) and anti-inflammatory effects. Retinol was incorporated into the nanoparticles to transport retinol-binding protein 4 (RBP4) to the kidneys, which abundantly express RBP receptors. The NLCs were fabricated by high-shear homogenization and sonication, and neutrophils were used as a model to assess their anti-inflammatory effects. Mice were injected with MRSA to establish a model of bacteremia with organ injury. Results The mean nanoparticle size and zeta potential of the NLCs were 171 nm and − 39 mV, respectively. Ciprofloxacin (0.05%, w/v) and rolipram (0.02%) achieved encapsulation percentages of 88% and 96%, respectively, in the nanosystems. The minimum bactericidal concentration of free ciprofloxacin against MRSA increased from 1.95 to 15.63 µg/ml when combined with rolipram, indicating a possible drug-drug interaction that reduced the antibacterial effect. Nanoparticle inclusion promoted the anti-MRSA activity of ciprofloxacin according to time-kill curves. The NLCs were found to be largely internalized into neutrophils and exhibited superior superoxide anion inhibition than free drugs. Retinol incorporation into the nanocarriers facilitated their efficient targeting to the kidneys. The NLCs significantly mitigated MRSA burden and elastase distribution in the organs of MRSA-infected animals, and the greatest inhibition was observed in the kidneys. Bacterial clearance and neutrophil infiltration suppression attenuated the bacteremia-induced cytokine overexpression, leading to an improvement in the survival rate from 22% to 67%. Conclusions The dual role of our NLCs endowed them with greater efficacy in treating MRSA bacteremia than that of free drugs. ![]()
Collapse
Affiliation(s)
- Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, 5 Fuxing Street, Kweishan, Taoyuan, 333, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, 5 Fuxing Street, Kweishan, Taoyuan, 333, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Shih-Chun Yang
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| | - Ahmed Alalaiwe
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - You-Shan Dai
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, 5 Fuxing Street, Kweishan, Taoyuan, 333, Taiwan. .,School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan.
| | - Jia-You Fang
- Department of Anesthesiology, Chang Gung Memorial Hospital, 5 Fuxing Street, Kweishan, Taoyuan, 333, Taiwan. .,Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan. .,Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|
192
|
Buccini DF, Cardoso MH, Franco OL. Antimicrobial Peptides and Cell-Penetrating Peptides for Treating Intracellular Bacterial Infections. Front Cell Infect Microbiol 2021; 10:612931. [PMID: 33614528 PMCID: PMC7892433 DOI: 10.3389/fcimb.2020.612931] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Bacterial infections caused by intracellular pathogens are difficult to control. Conventional antibiotic therapies are often ineffective, as high doses are needed to increase the number of antibiotics that will cross the host cell membrane to act on the intracellular bacterium. Moreover, higher doses of antibiotics may lead to elevated severe toxic effects against host cells. In this context, antimicrobial peptides (AMPs) and cell-penetrating peptides (CPPs) have shown great potential to treat such infections by acting directly on the intracellular pathogenic bacterium or performing the delivery of cargos with antibacterial activities. Therefore, in this mini-review, we cover the main AMPs and CPPs described to date, aiming at intracellular bacterial infection treatment. Moreover, we discuss some of the proposed mechanisms of action for these peptide classes and their conjugation with other antimicrobials.
Collapse
Affiliation(s)
- Danieli F Buccini
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Marlon H Cardoso
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Octavio L Franco
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| |
Collapse
|
193
|
Chain CY, Daza Millone MA, Cisneros JS, Ramirez EA, Vela ME. Surface Plasmon Resonance as a Characterization Tool for Lipid Nanoparticles Used in Drug Delivery. Front Chem 2021; 8:605307. [PMID: 33490037 PMCID: PMC7817952 DOI: 10.3389/fchem.2020.605307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/04/2020] [Indexed: 12/24/2022] Open
Abstract
The development of drug carriers based in lipid nanoparticles (LNPs) aims toward the synthesis of non-toxic multifunctional nanovehicles that can bypass the immune system and allow specific site targeting, controlled release and complete degradation of the carrier components. Among label free techniques, Surface Plasmon Resonance (SPR) biosensing is a versatile tool to study LNPs in the field of nanotherapeutics research. SPR, widely used for the analysis of molecular interactions, is based on the immobilization of one of the interacting partners to the sensor surface, which can be easily achieved in the case of LNPs by hydrophobic attachment onto commercial lipid- capture sensor chips. In the last years SPR technology has emerged as an interesting strategy for studying molecular aspects of drug delivery that determines the efficacy of the nanotherapeutical such as LNPs' interactions with biological targets, with serum proteins and with tumor extracelullar matrix. Moreover, SPR has contributed to the obtention and characterization of LNPs, gathering information about the interplay between components of the formulations, their response to organic molecules and, more recently, the quantification and molecular characterization of exosomes. By the combination of available sensor platforms, assay quickness and straight forward platform adaptation for new carrier systems, SPR is becoming a high throughput technique for LNPs' characterization and analysis.
Collapse
Affiliation(s)
- Cecilia Yamil Chain
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA- Universidad Nacional de La Plata (UNLP)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), La Plata, Argentina
| | - María Antonieta Daza Millone
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA- Universidad Nacional de La Plata (UNLP)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), La Plata, Argentina
| | - José Sebastián Cisneros
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA- Universidad Nacional de La Plata (UNLP)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), La Plata, Argentina
| | - Eduardo Alejandro Ramirez
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA- Universidad Nacional de La Plata (UNLP)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), La Plata, Argentina
| | - María Elena Vela
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA- Universidad Nacional de La Plata (UNLP)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)), La Plata, Argentina
| |
Collapse
|
194
|
Microwave assisted synthesis of Fe 3O 4 stabilized ZrO 2 nanoparticles - Free radical scavenging, radiolabeling and biodistribution in rabbits. Life Sci 2021; 271:119070. [PMID: 33465388 DOI: 10.1016/j.lfs.2021.119070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 11/24/2022]
Abstract
AIMS In vivo biodistribution of radio labeled ZrO2 nanoparticles is addressed for better imaging, therapy and diagnosis. Nanoparticles are synthesized by microwave assisted sol-gel technique using Fe3O4 as a stabilizer. Antioxidant assay, hemolytic activity in human blood and biodistribution in rabbits was explored to study the therapeutical as well as in vivo targeted diagnostic applications of as synthesized nanoparticles. MAIN METHODS Fe3O4 stabilized zirconia nanoparticles are synthesized using microwave assisted sol-gel method. Microwave (MW) powers are varied in the range of 100 to 1000 W. As synthesized nanoparticles are evaluated using different characterizations such as X-ray diffractometer, scanning electron microscope, Raman spectroscopy, impedance analyzer, Vickers micro hardness indenter, FTIR, and UV-Vis spectroscopy. In vitro activity of synthesized nanoparticles is checked in freshly extracted human blood serum. To study biodistribution of Fe3O4 stabilized zirconia nanoparticles in rabbit, technetium-99 m was used for labeling purpose. The labeling efficacy and stability of labeled nanoparticles are also measured with instant thin layer chromatography (ITLC) method. Intravenous injection of 99mTc-Fe3O4 stabilized zirconia nanoparticles (0.2 ml), containing 110 MBq of radioactivity, is performed to study the biodistribution; nanoparticles are injected into the ear vein of animal (rabbit). KEY FINDINGS Zirconia (ZrO2) nanoparticles (NPs) are stabilized using Fe3O4 that were prepared by means of microwave assisted sol-gel method. Crystallite size (~20 nm) agrees well with the values required to stabilize tetragonal zirconia (t-ZrO2). Volume shrinkage results in high value of hardness (~1369). Dielectric constant values, compatible for biomedical application, are observed for tetragonally stabilized samples. Low value of hemolytic response is observed for Fe3O4 stabilized ZrO2 NPs. 99mTc radio labeled ZrO2 NPs proved to be potential candidate to study biodistribution. Biodistribution studies show stability of radiolabeled NPs in the original suspension as well as in blood serum. CT scan of rabbit is performed for several times to check the biodistribution of NPs with time and survival of rabbit. Results suggest that these NPs can also be used as targeted nanoparticles as well as variants of drug payload carrier. SIGNIFICANCE Results signify that Fe3O4 stabilized ZrO2 nanoparticles synthesized by microwave assisted sol-gel method may be considered as "all-rounder" nanoplatform and are safe enough to be used in diagnostic as well as therapeutic purposes.
Collapse
|
195
|
Gupta P, Goel A, Singh KR, Meher MK, Gulati K, Poluri KM. Dissecting the anti-biofilm potency of kappa-carrageenan capped silver nanoparticles against Candida species. Int J Biol Macromol 2021; 172:30-40. [PMID: 33440209 DOI: 10.1016/j.ijbiomac.2021.01.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/31/2020] [Accepted: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Global antimicrobial crisis and advent of drug resistant fungal strains has substantially distressed disease management for clinicians. Biodegradable silver nanoparticles (AgNps) emerge as an excellent alternative remedial option. In the current study, the anti-biofilm activity of microwave irradiated kappa-carrageenan (CRG) capped AgNps against Candida albicans, and Candida glabrata was investigated in terms of their effect on reactive oxygen species (ROS) generation, cellular morphology, biochemical composition, and the activity of enzymes of extracellular matrix. Minimum inhibitory concentration and fungicidal concentration value of CRG-AgNps against both Candida spp. ranged between 400 and 500 μg/mL. The 80% of Candida biofilm was inhibited and eradicated by CRG-AgNps at a concentration of ~300 μg/mL. Microscopic studies indicate that CRG-AgNps caused morphological damage through membrane disruption and pore formation. Further, CRG-AgNps generated ROS in a concentration-dependent manner and modulated the composition of Candida biofilm ECM by increasing the carbohydrate and eDNA content. CRG-AgNps also significantly inactivated the hydrolytic enzymes, thus hindering the biofilm forming ability. In conclusion, all these results suggest that the CRG-AgNps are potential antifungal agents against Candida biofilms, and they inhibit/eradicate the fungal biofilms through multiple signalling mechanisms.
Collapse
Affiliation(s)
- Payal Gupta
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Apoorva Goel
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Khushboo Rani Singh
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Mukesh Kumar Meher
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Khushboo Gulati
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
196
|
Scutera S, Argenziano M, Sparti R, Bessone F, Bianco G, Bastiancich C, Castagnoli C, Stella M, Musso T, Cavalli R. Enhanced Antimicrobial and Antibiofilm Effect of New Colistin-Loaded Human Albumin Nanoparticles. Antibiotics (Basel) 2021; 10:57. [PMID: 33430076 PMCID: PMC7827731 DOI: 10.3390/antibiotics10010057] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/27/2022] Open
Abstract
Multidrug-resistant (MDR) Gram-negative bacteria (GNB), such as Acinetobacter and Klebsiella, are responsible for severe hospital-acquired infections. Colistin, despite its toxicity and low tissue penetration, is considered the last resort antibiotic against these microorganisms. Of concern, the use of Colistin has recently been compromised by the emergence of Colistin resistance. Herein, we developed a new formulation consisting of multifunctional chitosan-coated human albumin nanoparticles for the delivery of Colistin (Col/haNPs). Col/haNPs were in vitro characterized for encapsulation efficiency, drug release, stability and cytotoxicity and were evaluated for antibacterial activity against MDR GNB (Acinetobacter baumannii and Klebsiella pneumoniae). Col/haNPs showed sizes lower than 200 nm, high encapsulation efficiency (98.65%) and prolonged in vitro release of Colistin. The safety of the nanoformulation was demonstrated by a negligible cytotoxicity on human fibroblasts and hemolytic activity. Col/haNPs evidenced a high antibacterial effect with a significant decrease in MIC values compared to free Colistin, in particular against Col-resistant strains with a pronounced decline of bacterial growth over time. Moreover, Col/haNPs exhibited an inhibitory effect on biofilm formation that was 4 and 60 fold higher compared to free Colistin, respectively for Colistin susceptible and resistant A. baumannii. Our findings suggest that Col/haNPs could represent a promising Colistin nanocarrier with high antimicrobial activity on MDR GNB.
Collapse
Affiliation(s)
- Sara Scutera
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (S.S.); (R.S.)
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy; (M.A.); (F.B.); (C.B.); (R.C.)
| | - Rosaria Sparti
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (S.S.); (R.S.)
| | - Federica Bessone
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy; (M.A.); (F.B.); (C.B.); (R.C.)
| | - Gabriele Bianco
- Microbiology and Virology Unit, University Hospital Città della Salute e della Scienza di Torino, 10126 Turin, Italy;
| | - Chiara Bastiancich
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy; (M.A.); (F.B.); (C.B.); (R.C.)
- Institute Neurophysiopathol, INP, CNRS, Aix-Marseille University, 13005 Marseille, France
| | - Carlotta Castagnoli
- Skin Bank, Department of General and Specialized Surgery, University Hospital Città della Salute e della Scienza di Torino, 10126 Turin, Italy;
| | - Maurizio Stella
- Burn Center, CTO Hospital, Città della Salute e della Scienza di Torino, 10126 Turin, Italy;
| | - Tiziana Musso
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy; (S.S.); (R.S.)
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy; (M.A.); (F.B.); (C.B.); (R.C.)
| |
Collapse
|
197
|
Papafilippou L, Claxton A, Dark P, Kostarelos K, Hadjidemetriou M. Nanotools for Sepsis Diagnosis and Treatment. Adv Healthc Mater 2021; 10:e2001378. [PMID: 33236524 PMCID: PMC11469323 DOI: 10.1002/adhm.202001378] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Sepsis is one of the leading causes of death worldwide with high mortality rates and a pathological complexity hindering early and accurate diagnosis. Today, laboratory culture tests are the epitome of pathogen recognition in sepsis. However, their consistency remains an issue of controversy with false negative results often observed. Clinically used blood markers, C reactive protein (CRP) and procalcitonin (PCT) are indicators of an acute-phase response and thus lack specificity, offering limited diagnostic efficacy. In addition to poor diagnosis, inefficient drug delivery and the increasing prevalence of antibiotic-resistant microorganisms constitute significant barriers in antibiotic stewardship and impede effective therapy. These challenges have prompted the exploration for alternative strategies that pursue accurate diagnosis and effective treatment. Nanomaterials are examined for both diagnostic and therapeutic purposes in sepsis. The nanoparticle (NP)-enabled capture of sepsis causative agents and/or sepsis biomarkers in biofluids can revolutionize sepsis diagnosis. From the therapeutic point of view, currently existing nanoscale drug delivery systems have proven to be excellent allies in targeted therapy, while many other nanotherapeutic applications are envisioned. Herein, the most relevant applications of nanomedicine for the diagnosis, prognosis, and treatment of sepsis is reviewed, providing a critical assessment of their potentiality for clinical translation.
Collapse
Affiliation(s)
- Lana Papafilippou
- Nanomedicine LabFaculty of BiologyMedicine and HealthAV Hill BuildingThe University of ManchesterManchesterM13 9PTUK
| | - Andrew Claxton
- Department of Critical CareSalford Royal Foundation TrustStott LaneSalfordM6 8HDUK
| | - Paul Dark
- Manchester NIHR Biomedical Research CentreDivision of InfectionImmunity and Respiratory MedicineUniversity of ManchesterManchesterM13 9PTUK
| | - Kostas Kostarelos
- Nanomedicine LabFaculty of BiologyMedicine and HealthAV Hill BuildingThe University of ManchesterManchesterM13 9PTUK
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)Campus UABBellaterraBarcelona08193Spain
| | - Marilena Hadjidemetriou
- Nanomedicine LabFaculty of BiologyMedicine and HealthAV Hill BuildingThe University of ManchesterManchesterM13 9PTUK
| |
Collapse
|
198
|
Mela I, Kaminski CF. Nano-vehicles give new lease of life to existing antimicrobials. Emerg Top Life Sci 2020; 4:555-566. [PMID: 33258900 PMCID: PMC7752037 DOI: 10.1042/etls20200153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/04/2023]
Abstract
Antibiotic resistance has become one of the greatest challenges for modern medicine, and new approaches for the treatment of bacterial infections are urgently needed to avoid widespread vulnerability again to infections that have so far been easily treatable with existing drugs. Among the many approaches investigated to overcome this challenge is the use of engineered nanostructures for the precise and targeted delivery of existing antimicrobial agents in a fashion that will potentiate their effect. This idea leans on lessons learned from pioneering research in cancer, where the targeted delivery of anti-cancer drugs to mammalian cells has been a topic for some time. In particular, new research has demonstrated that nanomaterials can be functionalised with active antimicrobials and, in some cases, with targeting molecules that potentiate the efficiency of the antimicrobials. In this mini-review, we summarise results that demonstrate the potential for nanoparticles, dendrimers and DNA nanostructures for use in antimicrobial delivery. We consider material aspects of the delivery vehicles and ways in which they can be functionalised with antibiotics and antimicrobial peptides, and we review evidence for their efficacy to kill bacteria both in vitro and in vivo. We also discuss the advantages and limitations of these materials and highlight the benefits of DNA nanostructures specifically for their versatile potential in the present context.
Collapse
Affiliation(s)
- Ioanna Mela
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Clemens F Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| |
Collapse
|
199
|
Magnetic Nanoparticle-Based Drug Delivery Approaches for Preventing and Treating Biofilms in Cystic Fibrosis. MAGNETOCHEMISTRY 2020. [DOI: 10.3390/magnetochemistry6040072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biofilm-associated infections pose a huge burden on healthcare systems worldwide, with recurrent lung infections occurring due to the persistence of biofilm bacteria populations. In cystic fibrosis (CF), thick viscous mucus acts not only as a physical barrier, but also serves as a nidus for infection. Increased antibiotic resistance in the recent years indicates that current therapeutic strategies aimed at biofilm-associated infections are “failing”, emphasizing the need to develop new and improved drug delivery systems with higher efficacy and efficiency. Magnetic nanoparticles (MNPs) have unique and favourable properties encompassing biocompatibility, biodegradability, magnetic and heat-mediated characteristics, making them suitable drug carriers. Additionally, an external magnetic force can be applied to enhance drug delivery to target sites, acting as “nano-knives”, cutting through the bacterial biofilm layer and characteristically thick mucus in CF. In this review, we explore the multidisciplinary approach of using current and novel MNPs as vehicles of drug delivery. Although many of these offer exciting prospects for future biofilm therapeutics, there are also major challenges of this emerging field that need to be addressed.
Collapse
|
200
|
Chaves de Souza MP, de Mattos NH, Pedreiro LN, Boni FI, dos Santos Ramos MA, Bauab TM, Gremião MPD, Chorilli M. Design of Mucoadhesive Nanostructured Polyelectrolyte Complexes Based on Chitosan and Hypromellose Phthalate for Metronidazole Delivery Intended to the Treatment of Helicobacter pylori Infections. Pharmaceutics 2020; 12:E1211. [PMID: 33327588 PMCID: PMC7765050 DOI: 10.3390/pharmaceutics12121211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/06/2020] [Accepted: 12/10/2020] [Indexed: 01/13/2023] Open
Abstract
Metronidazole (MT) is an important drug available for Helicobacter pylori infection treatment. However, in the past few years, this drug has presented effective reduction for infection control, one of the most important reasons is attributed to the reduction of retention time in the stomach environment. Mucoadhesive nanostructured polyelectrolyte complexes (nano PECs) based on chitosan (CS) and hypromellose phthalate (HP) were rationally developed using a full factorial design (21 × 21 × 31), for the incorporation of MT based on the enhancement of the antimicrobial potential against active Helicobacter pylori, in the stomach. Different mass ratios of CS:HP (w/w) were tested, reaching the most promising ratios of 1:0.1, 1:0.5, and 1:1, and two methods of polymers addition (pouring-I and drip-II) were also evaluated. From method I, the obtained particles presented a diameter in the range of 811-1293 nm (Z-average) and a polydispersity index (PDI) between 0.47 and 0.88. By method II, there was a significant reduction in diameter and PDI to 553-739 nm and 0.23 at 0.34, respectively. The drug incorporation also resulted in a reduction in the diameter and PDI of the nano PECs. All samples showed positive zeta potential, about 20 mV, and a high percentage of MT incorporation (±95%). The method factor presented a greater influence on the nano PECs characteristics. Interactions in the system constituents were indicated by the FTIR data. Nano PECs mucoadhesiveness was observed and the composition and charge density were responsible for this phenomenon. MT dissolution evaluation showed the similarity of the dissolution profiles of free and loaded MT, in which almost 100% of the drug was in the simulated gastric medium in 120 min of testing. The in vitro antimicrobial potential against H. pylori of loaded nano PECs were measured and the minimum inhibitory concentration observed for free MT was >2000 µg/mL, while for the incorporated MT lower values were observed, showing an increase in the encapsulated MT activity.
Collapse
Affiliation(s)
- Maurício Palmeira Chaves de Souza
- Department of Drug and Medicines, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), Araraquara-SP, Road Araraquara-Jaú, Km 01, 14.800-903 São Paulo, Brazil; (N.H.d.M.); (L.N.P.); (F.I.B.)
| | - Nathalia Helena de Mattos
- Department of Drug and Medicines, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), Araraquara-SP, Road Araraquara-Jaú, Km 01, 14.800-903 São Paulo, Brazil; (N.H.d.M.); (L.N.P.); (F.I.B.)
| | - Liliane Neves Pedreiro
- Department of Drug and Medicines, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), Araraquara-SP, Road Araraquara-Jaú, Km 01, 14.800-903 São Paulo, Brazil; (N.H.d.M.); (L.N.P.); (F.I.B.)
| | - Fernanda Isadora Boni
- Department of Drug and Medicines, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), Araraquara-SP, Road Araraquara-Jaú, Km 01, 14.800-903 São Paulo, Brazil; (N.H.d.M.); (L.N.P.); (F.I.B.)
| | - Matheus Aparecido dos Santos Ramos
- Department of Biological Sciences, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), 14.800-903 São Paulo, Brazil; (M.A.d.S.R.); (T.M.B.)
| | - Taís Maria Bauab
- Department of Biological Sciences, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), 14.800-903 São Paulo, Brazil; (M.A.d.S.R.); (T.M.B.)
| | - Maria Palmira Daflon Gremião
- Department of Drug and Medicines, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), Araraquara-SP, Road Araraquara-Jaú, Km 01, 14.800-903 São Paulo, Brazil; (N.H.d.M.); (L.N.P.); (F.I.B.)
| | - Marlus Chorilli
- Department of Drug and Medicines, School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State University (UNESP), Araraquara-SP, Road Araraquara-Jaú, Km 01, 14.800-903 São Paulo, Brazil; (N.H.d.M.); (L.N.P.); (F.I.B.)
| |
Collapse
|