1
|
Pisani S, Tufail S, Rosalia M, Dorati R, Genta I, Chiesa E, Conti B. Antibiotic-Loaded Nano-Sized Delivery Systems: An Insight into Gentamicin and Vancomycin. J Funct Biomater 2024; 15:194. [PMID: 39057315 PMCID: PMC11277905 DOI: 10.3390/jfb15070194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The fight against infectious disease has remained an ever-evolving challenge in the landscape of healthcare. The ability of pathogens to develop resistance against conventional drug treatments has decreased the effectiveness of therapeutic interventions, and antibiotic resistance is recognized as one of the main challenges of our time. The goal of this systematic review paper is to provide insight into the research papers published on innovative nanosized drug delivery systems (DDSs) based on gentamycin and vancomycin and to discuss the opportunity of their repurposing through nano DDS formulations. These two antibiotics are selected because (i) gentamicin is the first-line drug used to treat suspected or confirmed infections caused by Gram-negative bacterial infections and (ii) vancomycin is used to treat serious Gram-positive bacterial infections. Moreover, both antibiotics have severe adverse effects, and one of the purposes of their formulation as nanosized DDSs is to overcome them. The review paper includes an introduction focusing on the challenges of infectious diseases and traditional therapeutic treatments, a brief description of the chemical and pharmacological properties of gentamicin and vancomycin, case studies from the literature on innovative nanosized DDSs as carriers of the two antibiotic drugs, and a discussion of the results found in the literature.
Collapse
Affiliation(s)
- Silvia Pisani
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (S.P.); (S.T.); (M.R.); (R.D.); (I.G.); (E.C.)
| | - Shafia Tufail
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (S.P.); (S.T.); (M.R.); (R.D.); (I.G.); (E.C.)
- Department of Drug Sciences, IUSS Scuola Universitaria Superiore Pavia, 27100 Pavia, Italy
| | - Mariella Rosalia
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (S.P.); (S.T.); (M.R.); (R.D.); (I.G.); (E.C.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (S.P.); (S.T.); (M.R.); (R.D.); (I.G.); (E.C.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (S.P.); (S.T.); (M.R.); (R.D.); (I.G.); (E.C.)
| | - Enrica Chiesa
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (S.P.); (S.T.); (M.R.); (R.D.); (I.G.); (E.C.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (S.P.); (S.T.); (M.R.); (R.D.); (I.G.); (E.C.)
| |
Collapse
|
2
|
de Almeida Campos LA, de Souza JB, de Queiroz Macêdo HLR, Borges JC, de Oliveira DN, Cavalcanti IMF. Synthesis of polymeric nanoparticles by double emulsion and pH-driven: encapsulation of antibiotics and natural products for combating Escherichia coli infections. Appl Microbiol Biotechnol 2024; 108:351. [PMID: 38819646 PMCID: PMC11142984 DOI: 10.1007/s00253-024-13114-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 06/01/2024]
Abstract
The design, development, and obtaining of nanostructured materials, such as polymeric nanoparticles, have garnered interest due to loading therapeutic agents and its broad applicability. Polymeric nanoparticle synthesis employs advanced techniques such as the double emulsion approach and the pH-driven method, allowing the efficient incorporation of active compounds into these matrices. These loading methods ensure compound stability within the polymeric structure and enable control of the release of therapeutic agents. The ability of loaded polymeric nanoparticles to transport and release therapeutic agents on target manner represents a significant advancement in the quest for effective therapeutic solutions. Amid escalating concerns regarding antimicrobial resistance, interventions using polymeric nanostructures stand out for the possibility of carrying antimicrobial agents and enhancing antibacterial action against antibiotic-resistant bacteria, making a new therapeutic approach or complement to conventional treatments. In this sense, the capability of these polymeric nanoparticles to act against Escherichia coli underscores their relevance in controlling bacterial infections. This mini-review provides a comprehensive synthesis of promising techniques for loading therapeutic agents into polymeric nanoparticles highlighting methodologies and their implications, addressing prospects of combating bacterial infections caused by E. coli. KEY POINTS: • The double emulsion method provides control over size and release of bioactives. • The pH-driven method improves the solubility, stability, and release of active. • The methods increase the antibacterial action of those encapsulated in PNPs.
Collapse
Affiliation(s)
- Luís André de Almeida Campos
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, Cidade Universitária, Recife, PE, 123550670-901, Brazil
| | - Jaqueline Barbosa de Souza
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, Cidade Universitária, Recife, PE, 123550670-901, Brazil
| | - Hanne Lazla Rafael de Queiroz Macêdo
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, Cidade Universitária, Recife, PE, 123550670-901, Brazil
| | - Joyce Cordeiro Borges
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, Cidade Universitária, Recife, PE, 123550670-901, Brazil
| | - David Nattan de Oliveira
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, Cidade Universitária, Recife, PE, 123550670-901, Brazil
| | - Isabella Macário Ferro Cavalcanti
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, Cidade Universitária, Recife, PE, 123550670-901, Brazil.
- Laboratory of Microbiology and Immunology, Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Vitória de Santo Antão, PE, Brazil.
| |
Collapse
|
3
|
Vanzan DF, Goma EP, Locatelli FR, Honorio TDS, Furtado PDS, Rodrigues CR, de Sousa VP, Mata dos Santos HA, do Carmo FA, Simon A, Pyrrho ADS, Ribeiro AJ, Cabral LM. Evaluation of Silybin Nanoparticles against Liver Damage in Murine Schistosomiasis mansoni Infection. Pharmaceutics 2024; 16:618. [PMID: 38794280 PMCID: PMC11125168 DOI: 10.3390/pharmaceutics16050618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Silybin (SIB) is a hepatoprotective drug known for its poor oral bioavailability, attributed to its classification as a class IV drug with significant metabolism during the first-pass effect. This study explored the potential of solid lipid nanoparticles with (SLN-SIB-U) or without (SLN-SIB) ursodeoxycholic acid and polymeric nanoparticles (PN-SIB) as delivery systems for SIB. The efficacy of these nanosystems was assessed through in vitro studies using the GRX and Caco-2 cell lines for permeability and proliferation assays, respectively, as well as in vivo experiments employing a murine model of Schistosomiasis mansoni infection in BALB/c mice. The mean diameter and encapsulation efficiency of the nanosystems were as follows: SLN-SIB (252.8 ± 4.4 nm, 90.28 ± 2.2%), SLN-SIB-U (252.9 ± 14.4 nm, 77.05 ± 2.8%), and PN-SIB (241.8 ± 4.1 nm, 98.0 ± 0.2%). In the proliferation assay with the GRX cell line, SLN-SIB and SLN-SIB-U exhibited inhibitory effects of 43.09 ± 5.74% and 38.78 ± 3.78%, respectively, compared to PN-SIB, which showed no inhibitory effect. Moreover, SLN-SIB-U demonstrated a greater apparent permeability coefficient (25.82 ± 2.2) than PN-SIB (20.76 ± 0.1), which was twice as high as that of SLN-SIB (11.32 ± 4.6) and pure SIB (11.28 ± 0.2). These findings suggest that solid lipid nanosystems hold promise for further in vivo investigations. In the murine model of acute-phase Schistosomiasis mansoni infection, both SLN-SIB and SLN-SIB-U displayed hepatoprotective effects, as evidenced by lower alanine amino transferase values (22.89 ± 1.6 and 23.93 ± 2.4 U/L, respectively) than those in control groups I (29.55 ± 0.7 U/L) and I+SIB (34.29 ± 0.3 U/L). Among the prepared nanosystems, SLN-SIB-U emerges as a promising candidate for enhancing the pharmacokinetic properties of SIB.
Collapse
Affiliation(s)
- Daniel Figueiredo Vanzan
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ester Puna Goma
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Fernanda Resende Locatelli
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Thiago da Silva Honorio
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Priscila de Souza Furtado
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Carlos Rangel Rodrigues
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Valeria Pereira de Sousa
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Hilton Antônio Mata dos Santos
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Flávia Almada do Carmo
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Alice Simon
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| | - Alexandre dos Santos Pyrrho
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - António José Ribeiro
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
- Group Genetics of Cognitive Dysfunction, I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4169-007 Porto, Portugal
| | - Lucio Mendes Cabral
- Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.F.V.); (F.A.d.C.)
| |
Collapse
|
4
|
Tong WY, Tan WN, Kamarul Azizi MA, Leong CR, El Azab IH, Lim JW, Mahmoud MHH, Dailin DJ, Ibrahim MM, Chuah LF. Nanoparticle-laden contact lens for controlled release of vancomycin with enhanced antibiotic efficacy. CHEMOSPHERE 2023; 338:139492. [PMID: 37451643 DOI: 10.1016/j.chemosphere.2023.139492] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Vancomycin is the last resort antibiotic for the treatment of severe bacterial keratitis. Its clinical application is limited due to its hydrophilicity and high molecular weight. To overcome this, this study aims to develop nanoparticles-laden contact lens for controlled ocular delivery of vancomycin. Polyvinyl alcohol (PVA) was used as encapsulant material. The nanoparticles had a negative surface charge and an average size of 147.6 nm. A satisfactory encapsulation efficiency (61.24%) was obtained. The release profile was observed to be slow and sustained, with a release rate of 1.29 μL mg-1 h-1 for 48 h. Five out of 6 test bacteria were suppressed by vancomycin nanoparticles-laden contact lens. Vancomycin is generally ineffective against Gram-negative bacteria and unable to pass through the outer membrane barrier. In this study, vancomycin inhibited Proteus mirabilis and Pseudomonas aeruginosa. Nano-encapsulation enables vancomycin to penetrate the Gram-negative cell wall and further destroy the bacterial cells. On Hohenstein challenge test, all test bacteria exhibited significant reduction in growth when exposed to vancomycin nanoparticles-laden contact lens. This study created an effective and long-lasting vancomycin delivery system via silicone hydrogel contact lenses, by using PVA as encapsulant. The antibiotic efficacy and vancomycin release should be further studied using ocular in vivo models.
Collapse
Affiliation(s)
- Woei Yenn Tong
- Universiti Kuala Lumpur, Branch Campus Institute of Medical Science Technology, A1, 1, Jalan TKS 1, Taman Kajang Sentral, 43000, Kajang, Selangor, Malaysia; Universiti Kuala Lumpur, Branch Campus Malaysian Institute of Chemical and Bioengineering Technology, Lot 1988 Kawasan Perindustrian Bandar Vendor, Taboh Naning, Alor Gajah, Melaka, Malaysia.
| | - Wen-Nee Tan
- School of Distance Education, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Muhammad Afif Kamarul Azizi
- Universiti Kuala Lumpur, Branch Campus Malaysian Institute of Chemical and Bioengineering Technology, Lot 1988 Kawasan Perindustrian Bandar Vendor, Taboh Naning, Alor Gajah, Melaka, Malaysia
| | - Chean Ring Leong
- Universiti Kuala Lumpur, Branch Campus Malaysian Institute of Chemical and Bioengineering Technology, Lot 1988 Kawasan Perindustrian Bandar Vendor, Taboh Naning, Alor Gajah, Melaka, Malaysia
| | - Islam H El Azab
- Department of Food Science and Nutrition, College of Science, Taif University, P.O. box 11099, Taif, 21944, Saudi Arabia
| | - Jun Wei Lim
- HICoE-Centre for Biofuel and Biochemical Research, Institute of Self Sustainable Building, Department of Fundamental and Applied Sciences, Universiti Teknologi PETRONAS, Seri Iskandar, 32610, Perak Darul Ridzuan, Malaysia; Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, India
| | - M H H Mahmoud
- Department of Chemistry, College of Science, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Daniel Joe Dailin
- Institute of Bioproduct Development, Universiti Teknologi Malaysia, Johor, Malaysia; Department of Bioprocess and Polymer Engineering, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, Johor, Malaysia.
| | - Mohamed M Ibrahim
- Department of Chemistry, College of Science, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Lai Fatt Chuah
- Faculty of Maritime Studies, Universiti Malaysia Terengganu, Terengganu, Malaysia.
| |
Collapse
|
5
|
de Souza Furtado P, Agnes Silva Camargo de Oliveira A, Santiago Rodrigues P, Rita Santiago de Paula Gonçalves A, Raphaella Autran Colaço A, Pinheiro da Costa S, Muniz da Paz M, Wetler Meireles Carreiros Assumpção P, Pereira Rangel L, Simon A, Almada do Carmo F, Mendes Cabral L, Cunha Sathler P. In vivo evaluation of time-dependent antithrombotic effect of rivaroxaban-loaded poly(lactic-co-glycolic acid)/sodium lauryl sulfate or didodecyl dimethylammonium bromide nanoparticles in Wistar rats. Eur J Pharm Biopharm 2023; 190:184-196. [PMID: 37517449 DOI: 10.1016/j.ejpb.2023.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Rivaroxaban (RVX), an oral direct factor Xa inhibitor, is being explored as an alternative to traditional anticoagulans. However, RVX still faces pharmacokinetic limitations and adverse effects, highlighting the need for more effective formulations. In this regard, pharmaceutical nanotechnology, particularly the use of polymeric nanoparticles (PNPs), offers a promising approach for optimizing RVX delivery. This study aimed to develop and physicochemically characterize RVX-loaded poly(lactic-co-glycolic acid) (PLGA)/sodium lauryl sulfate (SLS) or didodecyl dimethylammonium bromide (DMAB) nanoparticles, and also evaluate their pharmacological and toxicological profiles as a potential therapeutic strategy. The PNPs exhibited sizes below 300 nm and spherical morphology, with both negative and positive surface charges, according to surfactant used. They demonstrated high encapsulation efficiency and suitable yields, as well as rapid initial liberation followed by sustained release in different pH environments. Importantly, in vivo evaluations revealed a time-dependent antithrombotic effect surpassing the free form of RVX when administered orally in SLS or DMAB PNP. No hemolytic or cytotoxic effects were observed at various concentrations of the PNPs. Interestingly, the PNPs did not induce hemorrhagic events or cause liver enzyme alterations in vivo. These findings suggest that RVX-loaded SLS or DMAB PNPs are promising innovative therapeutic alternatives for the treatment of thromboembolic diseases.
Collapse
Affiliation(s)
- Priscila de Souza Furtado
- Universidade Federal do Rio de Janeiro, LabHEx, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | | | - Pryscila Santiago Rodrigues
- Universidade Federal do Rio de Janeiro, LabHEx, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | | | - Anna Raphaella Autran Colaço
- Universidade Federal do Rio de Janeiro, LabHEx, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | - Sandro Pinheiro da Costa
- Universidade Federal do Rio de Janeiro, LabHEx, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | - Mariana Muniz da Paz
- Universidade Federal do Rio de Janeiro, LBT, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | | | - Luciana Pereira Rangel
- Universidade Federal do Rio de Janeiro, LBT, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | - Alice Simon
- Universidade Federal do Rio de Janeiro, LabTIF, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | - Flávia Almada do Carmo
- Universidade Federal do Rio de Janeiro, LabTIF, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | - Lucio Mendes Cabral
- Universidade Federal do Rio de Janeiro, LabTIF, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil
| | - Plínio Cunha Sathler
- Universidade Federal do Rio de Janeiro, LabHEx, Faculdade de Farmácia, Ilha do Fundão, CEP 21941-902, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
6
|
Budama-Kilinc Y, Gok B, Cetin Aluc C, Kecel-Gunduz S. In vitro and in silico evaluation of the design of nano-phyto-drug candidate for oral use against Staphylococcus aureus. PeerJ 2023; 11:e15523. [PMID: 37309371 PMCID: PMC10257901 DOI: 10.7717/peerj.15523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/17/2023] [Indexed: 06/14/2023] Open
Abstract
Onopordum acanthium is a medicinal plant with many important properties, such as antibacterial, anticancer, and anti-hypotensive properties. Although various studies reported the biological activities of O. acanthium, there is no study on its nano-phyto-drug formulation. The aim of this study is to develop a candidate nano-drug based on phytotherapeutic constituents and evaluate its efficiency in vitro and in silico. In this context, poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) of O. acanthium extract (OAE) were synthesized and characterized. It was determined that the average particle size of OAE-PLGA-NPs was 214.9 ± 6.77 nm, and the zeta potential was -8.03 ± 0.85 mV, and PdI value was 0.064 ± 0.013. The encapsulation efficiency of OAE-PLGA-NPs was calculated as 91%, and the loading capacity as 75.83%. The in vitro drug release study showed that OAE was released from the PLGA NPs with 99.39% over the 6 days. Furthermore, the mutagenic and cytotoxic activity of free OAE and OAE-PLGA-NPs were evaluated by the Ames test and MTT test, respectively. Although 0.75 and 0.37 mg/mL free OAE concentrations caused both frameshift mutation and base pair substitution (p < 0.05), the administered OAE-PLGA NP concentrations were not mutagenic. It was determined with the MTT analysis that the doses of 0.75 and 1.5 mg/mL of free OAE had a cytotoxic effect on the L929 fibroblast cell line (p < 0.05), and OAE-PLGA-NPs had no cytotoxic effect. Moreover, the interaction between the OAE and S. aureus was also investigated using the molecular docking analysis method. The molecular docking and molecular dynamics (MD) results were implemented to elucidate the S. aureus MurE inhibition potential of OAE. It was shown that quercetin in the OAE content interacted significantly with the substantial residues in the catalytic pocket of the S. aureus MurE enzyme, and quercetin performed four hydrogen bond interactions corresponding to a low binding energy of -6.77 kcal/mol with catalytic pocket binding residues, which are crucial for the inhibition mechanism of S. aureus MurE. Finally, the bacterial inhibition values of free OAE and OAE-PLGA NPs were determined against S. aureus using a microdilution method. The antibacterial results showed that the inhibition value of the OAE-PLGA NPs was 69%. In conclusion, from the in vitro and in silico results of the nano-sized OAE-PLGA NP formulation produced in this study, it was evaluated that the formulation may be recommended as a safe and effective nano-phyto-drug candidate against S. aureus.
Collapse
Affiliation(s)
- Yasemin Budama-Kilinc
- Bioengineering Department, Yildiz Technical University, Istanbul, Turkey
- Health Biotechnology Joint Research and Application Center of Excellence, Istanbul, Turkey
| | - Bahar Gok
- Graduate School of Natural and Applied Science, Yildiz Technical University, Istanbul, Turkey
| | - Cigdem Cetin Aluc
- Graduate School of Natural and Applied Science, Yildiz Technical University, Istanbul, Turkey
- Abdi Ibrahim Production Facilities, Abdi Ibrahim Pharmaceuticals, Istanbul, Turkey
| | | |
Collapse
|
7
|
Application of Nanomaterials in the Prevention, Detection, and Treatment of Methicillin-Resistant Staphylococcus aureus (MRSA). Pharmaceutics 2022; 14:pharmaceutics14040805. [PMID: 35456638 PMCID: PMC9030647 DOI: 10.3390/pharmaceutics14040805] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 01/27/2023] Open
Abstract
Due to differences in geographic surveillance systems, chemical sanitization practices, and antibiotic stewardship (AS) implementation employed during the COVID-19 pandemic, many experts have expressed concerns regarding a future surge in global antimicrobial resistance (AMR). A potential beneficiary of these differences is the Gram-positive bacteria MRSA. MRSA is a bacterial pathogen with a high potential for mutational resistance, allowing it to engage various AMR mechanisms circumventing conventional antibiotic therapies and the host’s immune response. Coupled with a lack of novel FDA-approved antibiotics reaching the clinic, the onus is on researchers to develop alternative treatment tools to mitigate against an increase in pathogenic resistance. Mitigation strategies can take the form of synthetic or biomimetic nanomaterials/vesicles employed in vaccines, rapid diagnostics, antibiotic delivery, and nanotherapeutics. This review seeks to discuss the current potential of the aforementioned nanomaterials in detecting and treating MRSA.
Collapse
|
8
|
Singh S, Aldawsari HM, Alam A, Alqarni MHS, Ranjan S, Kesharwani P. Synthesis and antimicrobial activity of vancomycin–conjugated zinc coordination polymer nanoparticles against methicillin-resistant staphylococcus aureus. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
9
|
Berini F, Orlandi V, Gornati R, Bernardini G, Marinelli F. Nanoantibiotics to fight multidrug resistant infections by Gram-positive bacteria: hope or reality? Biotechnol Adv 2022; 57:107948. [PMID: 35337933 DOI: 10.1016/j.biotechadv.2022.107948] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/17/2022]
Abstract
The spread of antimicrobial resistance in Gram-positive pathogens represents a threat to human health. To counteract the current lack of novel antibiotics, alternative antibacterial treatments have been increasingly investigated. This review covers the last decade's developments in using nanoparticles as carriers for the two classes of frontline antibiotics active on multidrug-resistant Gram-positive pathogens, i.e., glycopeptide antibiotics and daptomycin. Most of the reviewed papers deal with vancomycin nanoformulations, being teicoplanin- and daptomycin-carrying nanosystems much less investigated. Special attention is addressed to nanoantibiotics used for contrasting biofilm-associated infections. The status of the art related to nanoantibiotic toxicity is critically reviewed.
Collapse
Affiliation(s)
- Francesca Berini
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Viviana Orlandi
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Rosalba Gornati
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Giovanni Bernardini
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| | - Flavia Marinelli
- Department of Biotechnology and Life Sciences, University of Insubria, via JH Dunant 3, 21100 Varese, Italy.
| |
Collapse
|
10
|
Yang D, Zheng X, Jiang L, Ye M, He X, Jin Y, Wu R. Functional Mapping of Phenotypic Plasticity of Staphylococcus aureus Under Vancomycin Pressure. Front Microbiol 2021; 12:696730. [PMID: 34566908 PMCID: PMC8458881 DOI: 10.3389/fmicb.2021.696730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
Phenotypic plasticity is the exhibition of various phenotypic traits produced by a single genotype in response to environmental changes, enabling organisms to adapt to environmental changes by maintaining growth and reproduction. Despite its significance in evolutionary studies, we still know little about the genetic control of phenotypic plasticity. In this study, we designed and conducted a genome-wide association study (GWAS) to reveal genetic architecture of how Staphylococcus aureus strains respond to increasing concentrations of vancomycin (0, 2, 4, and 6 μg/mL) in a time course. We implemented functional mapping, a dynamic model for genetic mapping using longitudinal data, to map specific loci that mediate the growth trajectories of abundance of vancomycin-exposed S. aureus strains. 78 significant single nucleotide polymorphisms were identified following analysis of the whole growth and development process, and seven genes might play a pivotal role in governing phenotypic plasticity to the pressure of vancomycin. These seven genes, SAOUHSC_00020 (walR), SAOUHSC_00176, SAOUHSC_00544 (sdrC), SAOUHSC_02998, SAOUHSC_00025, SAOUHSC_00169, and SAOUHSC_02023, were found to help S. aureus regulate antibiotic pressure. Our dynamic gene mapping technique provides a tool for dissecting the phenotypic plasticity mechanisms of S. aureus under vancomycin pressure, emphasizing the feasibility and potential of functional mapping in the study of bacterial phenotypic plasticity.
Collapse
Affiliation(s)
- Dengcheng Yang
- Center for Computational Biology, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Xuyang Zheng
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Libo Jiang
- Center for Computational Biology, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China.,College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Meixia Ye
- Center for Computational Biology, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China.,College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Xiaoqing He
- Center for Computational Biology, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China.,College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Yi Jin
- Center for Computational Biology, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China.,College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Rongling Wu
- Center for Computational Biology, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China.,College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China.,Department of Public Health Sciences and Statistics, Center for Statistical Genetics, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
11
|
Ndayishimiye J, Cao Y, Kumeria T, Blaskovich MAT, Falconer JR, Popat A. Engineering mesoporous silica nanoparticles towards oral delivery of vancomycin. J Mater Chem B 2021; 9:7145-7166. [PMID: 34525166 DOI: 10.1039/d1tb01430g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vancomycin (Van) is a key antibiotic of choice for the treatment of systemic methicillin resistant Staphylococcus aureus (MRSA) infections. However, due to its poor membrane permeability, it is administered parenterally, adding to the cost and effort of treatment. The poor oral bioavailability of Van is mainly due to its physico-chemical properties that limit its paracellular and transcellular transport across gastrointestinal (GI) epithelium. Herein we report the development of silica nanoparticles (SNPs)-based formulations that are able to enhance the epithelial permeability of Van. We synthesized SNPs of different pore sizes (2 nm and 9 nm) and modified their surface charge and polarity by attaching different functional groups (-NH2, -PO3, and -CH3). Van was loaded within these SNPs at a loading capacity in the range of ca. 18-29 wt%. The Van-loaded SNPs exhibited a controlled release behaviour when compared to un-encapsulated Van which showed rapid release due to its hydrophilic nature. Among Van-loaded SNPs, SNPs with large pores showed a prolonged release compared to SNPs with small pores while SNPs functionalised with -CH3 groups exhibited a slowest release among the functionalised SNPs. Importantly, Van-loaded SNPs, especially the large pore SNPs with negative charge, enhanced the permeability of Van across an epithelial cell monolayer (Caco-2 cell model) by up to 6-fold, with Papp values up to 1.716 × 10-5 cm s-1 (vs. 0.304 × 10-5 cm s-1 for un-encapsulated Van) after 3 h. The enhancement was dependent on both the type of SNPs and their surface functionalisation. The permeation enhancing effect of SNPs was due to its ability to transiently open the tight junctions measured by decrease in transepithelial resistance (TEER) which was reversible after 3 h. All in all, our data highlights the potential of SNPs (especially SNPs with large pores) for oral delivery of Van or other antimicrobial peptides.
Collapse
Affiliation(s)
- John Ndayishimiye
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, Queensland 4102, Australia.
| | - Yuxue Cao
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, Queensland 4102, Australia.
| | - Tushar Kumeria
- School of Materials Science and Engineering, University of New South Wales, New South Wales, Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - James Robert Falconer
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, Queensland 4102, Australia.
| | - Amirali Popat
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, Queensland 4102, Australia. .,Mater Research Institute - The University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
12
|
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
|
13
|
Machado ME, de Souza Furtado P, da Costa Bernardes Araújo C, Simon A, de Moraes MC, Rodrigues Pereira da Silva LC, do Carmo FA, Cabral LM, Sathler PC. Novel rivaroxaban-loaded poly(lactic-co-glycolic acid)/poloxamer nanoparticles: preparation, physicochemical characterization, in vitro evaluation of time-dependent anticoagulant activity and toxicological profile. NANOTECHNOLOGY 2021; 32:135101. [PMID: 33276347 DOI: 10.1088/1361-6528/abd0b5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Rivaroxaban (RXB), an oral direct factor Xa inhibitor, presents innovative therapeutic profile. However, RXB has shown adverse effects, mainly due to pharmacokinetic limitations, highlighting the importance of developing more effective formulations. Therefore, this work aims at the preparation, physicochemical characterization and in vitro evaluation of time-dependent anticoagulant activity and toxicology profile of RXB-loaded poly(lactic-co-glycolic acid) (PLGA)/poloxamer nanoparticles (RXBNps). RXBNp were produced by nanoprecipitation method and physicochemical characteristics were evaluated. In vitro analysis of time-dependent anticoagulant activity was performed by prothrombin time test and toxicological profile was assessed by hemolysis and MTT reduction assays. The developed RXBNp present spherical morphology with average diameter of 205.5 ± 16.95 nm (PdI 0.096 ± 0.04), negative zeta potential (-26.28 ± 0.77 mV), entrapment efficiency of 91.35 ± 2.40%, yield of 41.81 ± 1.68% and 3.72 ± 0.07% of drug loading. Drug release was characterized by an initial fast release followed by a sustained release with 28.34 ± 2.82% of RXB available in 72 h. RXBNp showed an expressive time-dependent anticoagulant activity in human and rat blood plasma and non-toxic profile. Based on the results presented, it is possible to consider that RXBNp may be able to assist in the development of promising new therapies for treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Monique Etnea Machado
- Federal University of Rio de Janeiro, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco A Subsolo, sl24, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| | - Priscila de Souza Furtado
- Federal University of Rio de Janeiro, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco A Subsolo, sl24, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| | - Cristina da Costa Bernardes Araújo
- Federal University of Rio de Janeiro, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco A Subsolo, sl24, Rio de Janeiro, RJ, CEP 21941-902, Brazil
- Federal University of Rio de Janeiro, Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco L Subsolo, sl20, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| | - Alice Simon
- Federal University of Rio de Janeiro, Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco L Subsolo, sl20, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| | - Marcela Cristina de Moraes
- Fluminense Federal University, Department of Organic Chemistry, Outeiro de São João Batista s/n, Niterói, RJ, CEP 24210-240, Brazil
| | - Luiz Cláudio Rodrigues Pereira da Silva
- Federal University of Rio de Janeiro, Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco L Subsolo, sl20, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| | - Flávia Almada do Carmo
- Federal University of Rio de Janeiro, Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco L Subsolo, sl20, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| | - Lucio Mendes Cabral
- Federal University of Rio de Janeiro, Department of Drugs and Pharmaceutics, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco L Subsolo, sl20, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| | - Plínio Cunha Sathler
- Federal University of Rio de Janeiro, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Av. Carlos Chagas Filho, 373, CCS, Bloco A Subsolo, sl24, Rio de Janeiro, RJ, CEP 21941-902, Brazil
| |
Collapse
|
14
|
Walduck A, Sangwan P, Vo QA, Ratcliffe J, White J, Muir BW, Tran N. Treatment of Staphylococcus aureus skin infection in vivo using rifampicin loaded lipid nanoparticles. RSC Adv 2020; 10:33608-33619. [PMID: 35515067 PMCID: PMC9056717 DOI: 10.1039/d0ra06120d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
We have previously reported on a novel nanoparticle formulation that was effective at killing Staphylococcus aureus in vitro. Here, we report for the first time, the antibacterial effects of a lipidic nano-carrier containing rifampicin (NanoRIF) which can be used to successfully treat Methicillin-Resistant S. aureus (MRSA) infection at a reduced antibiotic dosage compared to the free drug in a skin wound model in mice. The formulation used contains the lipid monoolein, a cationic lipid N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl-sulfate (DOTAP) and the antibiotic. We have shown that rifampicin-loaded nanoparticles are more effective at treating infection in the skin wound model than the antibiotic alone. Cryo-TEM was used to capture for the first time, interactions of the formed nanoparticles with the cell wall of an individual bacterium. Our data strongly indicate enhanced binding of these charged nanoparticles with the negatively charged bacterial membrane. The efficacy we have now observed in vivo is of significant importance for the continued development of nanomedicine-based strategies to combat antibiotic resistant bacterial skin infections.
Collapse
Affiliation(s)
- Anna Walduck
- School of Science, RMIT University 124 La Trobe Street Melbourne 3000 Victoria Australia
| | - Parveen Sangwan
- CSIRO Manufacturing Bag 10 Clayton South 3169 Victoria Australia
| | - Quynh Anh Vo
- CSIRO Manufacturing Bag 10 Clayton South 3169 Victoria Australia
- Chimie Paris Tech Paris France
| | - Julian Ratcliffe
- CSIRO Manufacturing Bag 10 Clayton South 3169 Victoria Australia
| | - Jacinta White
- CSIRO Manufacturing Bag 10 Clayton South 3169 Victoria Australia
| | - Benjamin W Muir
- CSIRO Manufacturing Bag 10 Clayton South 3169 Victoria Australia
| | - Nhiem Tran
- School of Science, RMIT University 124 La Trobe Street Melbourne 3000 Victoria Australia
| |
Collapse
|