1
|
Elhassan E, Omolo CA, Gafar MA, Ismail EA, Ibrahim UH, Khan R, Lesouhaitier M, Kubes P, Govender T. Multifunctional hyaluronic acid-based biomimetic/pH-responsive hybrid nanostructured lipid carriers for treating bacterial sepsis. J Biomed Sci 2025; 32:19. [PMID: 39930418 PMCID: PMC11812216 DOI: 10.1186/s12929-024-01114-6] [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: 07/12/2024] [Accepted: 12/17/2024] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION The application of biomimetic and stimuli-responsive nanocarriers displays considerable promise in improving the management of bacterial sepsis and overcoming antimicrobial resistance. Therefore, the study aimed to synthesize a novel hyaluronic acid-lysine conjugate (HA-Lys) and to utilize the attributes of the synthesized HA-Lys with Tocopherol succinate (TS) and Oleylamine (OLA) in the formulation of multifunctional biomimetic pH-responsive HNLCs loaded with vancomycin (VCM-HNLCs), to combat bacterial sepsis. METHODS A novel hyaluronic acid-lysine conjugate (HA-Lys) was synthesized and characterized using FTIR and 1H NMR spectroscopy. Vancomycin-loaded hybrid nanosystems (VCM-HNLCs) were prepared through hot homogenization ultrasonication and evaluated for particle size, polydispersity index (PDI), zeta potential (ZP), and encapsulation efficiency (EE%). In vitro biocompatibility was assessed via MTT assay and red blood cell hemolysis test. The binding affinity to TLR2 and TLR4 was measured using microscale thermophoresis (MST). Drug release was evaluated using the dialysis bag method. Antimicrobial activity against MRSA and efflux pump inhibition were also determined. Efficacy was demonstrated in an MRSA-induced sepsis mice model. RESULTS The VCM-HNLCs, produced via hot homogenization ultrasonication, exhibited particle size (PS), polydispersity index (PDI), zeta potential (ZP), and encapsulation efficiency (EE%) of 110.77 ± 1.692 nm, 0.113 ± 0.022, - 2.92 ± 0.210 mV, and 76.27 ± 1.200%, respectively. In vitro, biocompatibility was proven by hemolysis and cytotoxicity studies. The VCM-HNLCs demonstrated targetability to human Toll-like receptors (TLR 2 and 4) as validated by microscale thermophoresis (MST). VCM-HNLCs showed a twofold reduction in MIC values at physiological pH compared to the bare VCM against S. aureus and MRSA for 48 h. While at pH 6.0, MIC values were reduced by fourfold in the first 24 h and by eightfold in the subsequent 48 and 72 h against tested strains. Furthermore, VCM-HNLCs showed inhibitory effects against MRSA efflux pumps, reactive oxygen species (ROS), and lipopolysaccharide (LPS)-induced hyperinflammation. In an MRSA-induced sepsis mice model, VCM-HNLCs demonstrated superior efficacy compared to free VCM, significantly eliminated bacteria and improved survival rates. CONCLUSIONS Overall, these results highlight the potential of VCM-HNLCs as novel multifunctional nanocarriers to combat antimicrobial resistance (AMR) and enhance sepsis outcomes.
Collapse
Affiliation(s)
- Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P. O. Box 14634-00800, Nairobi, Kenya.
| | - Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Medani, Sudan
| | - Usri H Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Rene Khan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Mathieu Lesouhaitier
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AL, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AL, Canada
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| |
Collapse
|
2
|
Bian X, Zhou L, Luo Z, Liu G, Hang Z, Li H, Li F, Wen Y. Emerging Delivery Systems for Enabling Precision Nucleic Acid Therapeutics. ACS NANO 2025; 19:4039-4083. [PMID: 39834294 DOI: 10.1021/acsnano.4c11858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Nucleic acid therapeutics represent a highly promising treatment approach in modern medicine, treating diseases at the genetic level. However, these therapeutics face numerous challenges in practical applications, particularly regarding their stability, effectiveness, cellular uptake efficiency, and limitations in delivering them specifically to target tissues. To overcome these obstacles, researchers have developed various innovative delivery systems, including viral vectors, lipid nanoparticles, polymer nanoparticles, inorganic nanoparticles, protein carriers, exosomes, antibody oligonucleotide conjugates, and DNA nanostructure-based delivery systems. These systems enhance the therapeutic efficacy of nucleic acid drugs by improving their stability, targeting specificity, and half-life in vivo. In this review, we systematically discuss different types of nucleic acid drugs, analyze the major barriers encountered in their delivery, and summarize the current research progress in emerging delivery systems. We also highlight the latest advancements in the application of these systems for treating genetic diseases, infectious diseases, cancer, brain diseases, and wound healing. This review aims to provide a comprehensive overview of nucleic acid drug delivery systems' current status and future directions by integrating the latest advancements in nanotechnology, biomaterials science, and gene editing technologies, emphasizing their transformative potential in precision medicine.
Collapse
Affiliation(s)
- Xiaochun Bian
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhiwei Luo
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Guotao Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhongci Hang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Haohao Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Fengyong Li
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
3
|
Wang Y, Yang J, Amier Y, Yuan D, Xun Y, Yu X. Advancements in Nanomedicine for the Diagnosis and Treatment of Kidney Stones. Int J Nanomedicine 2025; 20:1401-1423. [PMID: 39925679 PMCID: PMC11805677 DOI: 10.2147/ijn.s504318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/17/2025] [Indexed: 02/11/2025] Open
Abstract
Kidney stones constitute a common condition impacting the urinary system. In clinical diagnosis and management, traditional surgical interventions and pharmacological treatments are primarily utilized; however, these methods possess inherent limitations. Presently, the field of nanomedicine is undergoing significant advancements. The application of nanomaterials in biosensors enables the accurate assessment of urinary ion composition. Furthermore, contrast agents developed from these materials can improve the signal-to-noise ratio and enhance image clarity. By mitigating oxidative stress-induced cellular damage, nanomaterials can inhibit the formation of kidney stones and enhance the efficacy of drug delivery as effective carriers. Additionally, by modifying the physical and chemical properties of bacteria, nanomaterials can effectively eliminate bacterial presence, thereby preventing severe complications. This review explores the advancements in nanomaterials technology related to the early detection of risk factors, clinical diagnosis, and treatment of kidney stones and their associated complications.
Collapse
Affiliation(s)
- Yongqi Wang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Junyi Yang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yirixiatijiang Amier
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Dongfeng Yuan
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yang Xun
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiao Yu
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
4
|
Vaziri M, Abedini Baghbadorani M, Khandaee Ghamsari M, Handali S. The prospect of using nanotechnology to prevent and treat infections caused by Listeria monocytogenes. Crit Rev Microbiol 2025:1-9. [PMID: 39810637 DOI: 10.1080/1040841x.2025.2452571] [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: 09/07/2023] [Revised: 10/28/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Listeria monocytogenes (L. monocytogenes) is an opportunistic intracellular pathogen that causes listeriosis in human and leads to high mortality rate. L. monocytogenes is resistant to various antibiotics due to its ability to form biofilm. Designing a new generation of antibiotics is a very expensive and time-consuming process. Moreover, the protection of antibiotics via drug delivery system can promote their effectiveness and bioavailability. Nanomedicine can be a promising tool for treating intracellular bacteria and preventing the recurrence of infections. Nanocarriers can be employed as antibacterial agents or as a carrier for antibacterial agents. In the present review, the application of nanotechnology has been discussed for the prevention and treatment of Listeria infection. According to the studies, the application of nanomaterials can be a potential strategy to eradicate infections caused by L. monocytogenes.
Collapse
Affiliation(s)
- Mohammad Vaziri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Monireh Khandaee Ghamsari
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Zang J, Zhang L, Guo R, Kong L, Yu Y, Li S, Liu M, Wang J, Zhang Z, Li X, Liu Y. Baicalein loaded liposome with hyaluronic acid and Polyhexamethylene guanidine modification for anti methicillin-resistant Staphylococcus aureus infection. Int J Biol Macromol 2024; 276:133432. [PMID: 38936579 DOI: 10.1016/j.ijbiomac.2024.133432] [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: 12/17/2023] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Targeting delivery to the infection site and good affinity of vehicle to the bacterial are two main concerns in therapy of bacterial infection, and on-demand release of drug is another important issue. In this work, a liposome drug delivery system (HA/P/BAI-lip) incorporated with baicalein and modified by PHMG and HA was prepared. Several characterizations were conducted to examine the physical properties of liposome. Then it was applied to treatments of MRSA induced dorsal subcutaneous abscess model and the thigh muscle infected model. The presence of guanidine group in HA/P/BAI-lip rendered the liposome satisfactory bacterial target ability and good pH sensitive properties. The lipase secreted by bacterial could promote the hydrolysis of soybean phosphatidylcholine (SPC) in liposome. The modification of HA in HA/P/BAI-lip could lead the drug system to the exact infected site where CD44 was abundant because of inflammation. The low pH microenvironment characteristic of bacterial infection could induce the swelling of liposome following by degradation. Taken together, baicalein could be released selectively at the infected site to exert antibacterial capacity. HA/P/BAI-lip showed impressive antibacterial ability and dramatically decrease the bacterial burden of infection site and alleviate the infiltration of inflammatory cells, facilitating the recovery of infection.
Collapse
Affiliation(s)
- Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Shutong Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Mo Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Jiahua Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Zixu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Xuetao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China.
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China.
| |
Collapse
|
6
|
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
|
7
|
Dai X, Li Y, Liu X, Zhang Y, Gao F. Intracellular infection-responsive macrophage-targeted nanoparticles for synergistic antibiotic immunotherapy of bacterial infection. J Mater Chem B 2024; 12:5248-5260. [PMID: 38712662 DOI: 10.1039/d4tb00409d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Intracellular bacteria are considered to play a key role in the failure of bacterial infection therapy and increase of antibiotic resistance. Nanotechnology-based drug delivery carriers have been receiving increasing attention for improving the intracellular antibacterial activity of antibiotics, but are accompanied by disadvantages such as complex preparation procedures, lack of active targeting, and monotherapy, necessitating further design improvements. Herein, nanoparticles targeting bacteria-infected macrophages are fabricated to eliminate intracellular bacterial infections via antibiotic release and upregulation of intracellular reactive oxygen species (ROS) levels and proinflammatory responses. These nanoparticles were formed through the reaction of the amino group on selenocystamine dihydrochloride and the aldehyde group on oxidized dextran (ox-Dex), which encapsulates vancomycin (Van) through hydrophobic interactions. These nanoparticles could undergo targeted uptake by macrophages via endocytosis and respond to the bacteria-infected intracellular microenvironment (ROS and glutathione (GSH)) for controlled release of antibiotics. Furthermore, these nanoparticles could consume intracellular GSH and promote a significant increase in the level of ROS in macrophages, subsequently up-regulating the proinflammatory response to reinforce antibacterial activity. These nanoparticles can accelerate bacteria-infected wound healing. In this work, nanoparticles were fabricated for bacteria-infected macrophage-targeted and microenvironment-responsive antibiotic delivery, cellular ROS generation, and proinflammatory up-regulation activity to eliminate intracellular bacteria, which opens up a new possibility for multifunctional drug delivery against intracellular infection.
Collapse
Affiliation(s)
- Xiaomei Dai
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China.
| | - Yu Li
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China.
| | - Xiaojun Liu
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China.
| | - Yongjie Zhang
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China.
| | - Feng Gao
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China.
| |
Collapse
|
8
|
Dai X, Liu X, Wang X, Zhang Y, Li Y, Gao F. Cascade-Targeted Nanoplatforms for Synergetic Antibiotic/ROS/NO/Immunotherapy against Intracellular Bacterial Infection. Biomacromolecules 2024; 25:3190-3199. [PMID: 38693753 DOI: 10.1021/acs.biomac.4c00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Intracellular bacteria in dormant states can escape the immune response and tolerate high-dose antibiotic treatment, leading to severe infections. To overcome this challenge, cascade-targeted nanoplatforms that can target macrophages and intracellular bacteria, exhibiting synergetic antibiotic/reactive oxygen species (ROS)/nitric oxide (NO)/immunotherapy, were developed. These nanoplatforms were fabricated by encapsulating trehalose (Tr) and vancomycin (Van) into phosphatidylserine (PS)-coated poly[(4-allylcarbamoylphenylboric acid)-ran-(arginine-methacrylamide)-ran-(N,N'-bisacryloylcystamine)] nanoparticles (PABS), denoted as PTVP. PS on PTVP simulates a signal of "eat me" to macrophages to promote cell uptake (the first-step targeting). After the uptake, the nanoplatform in the acidic phagolysosomes could release Tr, and the exposed phenylboronic acid on the nanoplatform could target bacteria (the second-step targeting). Nanoplatforms can release Van in response to infected intracellular overexpressed glutathione (GSH) and weak acid microenvironment. l-arginine (Arg) on the nanoplatforms could be catalyzed by upregulated inducible nitric oxide synthase (iNOS) in the infected macrophages to generate nitric oxide (NO). N,N'-Bisacryloylcystamine (BAC) on nanoplatforms could deplete GSH, allow the generation of ROS in macrophages, and then upregulate proinflammatory activity, leading to the reinforced antibacterial capacity. This nanoplatform possesses macrophage and bacteria-targeting antibiotic delivery, intracellular ROS, and NO generation, and pro-inflammatory activities (immunotherapy) provides a new strategy for eradicating intracellular bacterial infections.
Collapse
Affiliation(s)
- Xiaomei Dai
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China
| | - Xiaojun Liu
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China
| | - Xingxing Wang
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China
| | - Yongjie Zhang
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China
| | - Yu Li
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China
| | - Feng Gao
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Province Key Laboratory of Biomedical Materials and Chemical Measurement, College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China
| |
Collapse
|
9
|
Tu YC, Wang YM, Yao LJ. Macrophage-Targeting DNA Nanomaterials: A Future Direction of Biological Therapy. Int J Nanomedicine 2024; 19:3641-3655. [PMID: 38681094 PMCID: PMC11055528 DOI: 10.2147/ijn.s459288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
DNA can be used for precise construction of complex and flexible micro-nanostructures, including DNA origami, frame nucleic acids, and DNA hydrogels. DNA nanomaterials have good biocompatibility and can enter macrophages via scavenger receptor-mediated endocytosis. DNA nanomaterials can be uniquely and flexibly designed to ensure efficient uptake by macrophages, which represents a novel strategy to regulate macrophage function. With the development of nanotechnology, major advances have been made in the design and manufacturing of DNA nanomaterials for clinical therapy. In diseases accompanied by macrophage disturbances including tumor, infectious diseases, arthritis, fibrosis, acute lung injury, and atherosclerosis, DNA nanomaterials received considerable attention as potential treatments. However, we lack sufficient information to guarantee precise targeting of macrophages by DNA nanomaterials, which precludes their therapeutic applications. In this review, we summarize recent studies of macrophage-targeting DNA nanomaterials and discuss the limitations and challenges of this approach with regard to its potential use as a biological therapy.
Collapse
Affiliation(s)
- Yu-Chi Tu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yu-Mei Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Li-Jun Yao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
10
|
Das S, Tinguely JC, Obuobi SAO, Škalko-Basnet N, Saxena K, Ahluwalia BS, Mehta DS. Plasmonic nano-bowls for monitoring intra-membrane changes in liposomes, and DNA-based nanocarriers in suspension. BIOMEDICAL OPTICS EXPRESS 2024; 15:2293-2307. [PMID: 38633091 PMCID: PMC11019686 DOI: 10.1364/boe.517471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/10/2024] [Accepted: 02/10/2024] [Indexed: 04/19/2024]
Abstract
Programmable nanoscale carriers, such as liposomes and DNA, are readily being explored for personalized medicine or disease prediction and diagnostics. The characterization of these nanocarriers is limited and challenging due to their complex chemical composition. Here, we demonstrate the utilization of surface-enhanced Raman spectroscopy (SERS), which provides a unique molecular fingerprint of the analytes while reducing the detection limit. In this paper, we utilize a silver coated nano-bowl shaped polydimethylsiloxane (PDMS) SERS substrate. The utilization of nano-bowl surface topology enabled the passive trapping of particles by reducing mobility, which results in reproducible SERS signal enhancement. The biological nanoparticles' dwell time in the nano-trap was in the order of minutes, thus allowing SERS spectra to remain in their natural aqueous medium without the need for drying. First, the geometry of the nano-traps was designed considering nanosized bioparticles of 50-150 nm diameter. Further, the systematic investigation of maximum SERS activity was performed using rhodamine 6 G as a probe molecule. The potential of the optimized SERS nano-bowl is shown through distinct spectral features following surface- (polyethylene glycol) and bilayer- (cholesterol) modification of empty liposomes of around 140 nm diameter. Apart from liposomes, the characterization of the highly crosslinked DNA specimens of only 60 nm in diameter was performed. The modification of DNA gel by liposome coating exhibited unique signatures for nitrogenous bases, sugar, and phosphate groups. Further, the unique sensitivity of the proposed SERS substrate displayed distinct spectral signatures for DNA micelles and drug-loaded DNA micelles, carrying valuable information to monitor drug release. In conclusion, the findings of the spectral signatures of a wide range of molecular complexes and chemical morphology of intra-membranes in their natural state highlight the possibilities of using SERS as a sensitive and instantaneous characterization alternative.
Collapse
Affiliation(s)
- Sathi Das
- Bio-photonics and Green Photonics Laboratory, Indian Institute of Technology Delhi, Hauz-Khas, New Delhi 110016, India
| | - Jean-Claude Tinguely
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Sybil Akua Okyerewa Obuobi
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway
| | - Kanchan Saxena
- Amity Institute of Renewable and Alternative Energy, Amity University Uttar Pradesh, Sector 125 Noida, U.P., India
| | - Balpreet Singh Ahluwalia
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway
| | - Dalip Singh Mehta
- Bio-photonics and Green Photonics Laboratory, Indian Institute of Technology Delhi, Hauz-Khas, New Delhi 110016, India
| |
Collapse
|
11
|
Li F, Cao D, Yao L, Gu W, Liu Z, Li D, Cui L. Targeted delivery of miR-34a-5p by phenylborate-coupled polyethylenimide nanocarriers for anti-KSHV treatment. Front Bioeng Biotechnol 2024; 11:1343956. [PMID: 38260739 PMCID: PMC10801047 DOI: 10.3389/fbioe.2023.1343956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) can infect a variety of cells and cause malignant tumors. At present, the use of microRNA (miRNA) for anti-KSHV is a promising treatment strategy, but the instability and non-specific uptake of miRNA still limit its use in the treatment of KSHV. In the present study, we constructed a nano-drug delivery system employing chemical grafting and electrostatic adsorption to solve the problems of easy degradation and low cell uptake of miRNA during direct administration. This nano-drug delivery system is to graft 4-carboxyphenylboric acid (PBA) and lauric acid (LA) onto polyethylenimine (PEI) through amidation reaction, and then prepare cationic copolymer nanocarriers (LA-PEI-PBA). The drug-carrying nanocomplex LA-PEI-PBA/miR-34a-5p was formed after further electrostatic adsorption of miR-34a-5p on the carrier and could protect miR-34a-5p from nuclease and serum degradation. Modification of the drug-carrying nanocomplex LA-PEI-PBA/miR-34a-5p by targeted molecule PBA showed effective uptake, increase in the level of miR-34a-5p, and inhibition of cell proliferation and migration in KSHV-infected cells. In addition, the drug-carrying nanocomplex could also significantly reduce the expression of KSHV lytic and latent genes, achieving the purpose of anti-KSHV treatment. In conclusion, these cationic copolymer nanocarriers with PBA targeting possess potential applications in nucleic acid delivery and anti-KSHV therapy.
Collapse
Affiliation(s)
- Fangling Li
- School of Chemistry and Chemical Engineering, State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi, Xinjiang, China
| | - Dongdong Cao
- School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Lixia Yao
- School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland (UQ), Brisbane, QLD, Australia
| | - Zhiyong Liu
- School of Chemistry and Chemical Engineering, State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi, Xinjiang, China
| | - Dongmei Li
- School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Lin Cui
- School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
12
|
Kamankesh M, Yadegar A, Llopis-Lorente A, Liu C, Haririan I, Aghdaei HA, Shokrgozar MA, Zali MR, Miri AH, Rad-Malekshahi M, Hamblin MR, Wacker MG. Future Nanotechnology-Based Strategies for Improved Management of Helicobacter pylori Infection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302532. [PMID: 37697021 DOI: 10.1002/smll.202302532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/25/2023] [Indexed: 09/13/2023]
Abstract
Helicobacter pylori (H. pylori) is a recalcitrant pathogen, which can cause gastric disorders. During the past decades, polypharmacy-based regimens, such as triple and quadruple therapies have been widely used against H. pylori. However, polyantibiotic therapies can disturb the host gastric/gut microbiota and lead to antibiotic resistance. Thus, simpler but more effective approaches should be developed. Here, some recent advances in nanostructured drug delivery systems to treat H. pylori infection are summarized. Also, for the first time, a drug release paradigm is proposed to prevent H. pylori antibiotic resistance along with an IVIVC model in order to connect the drug release profile with a reduction in bacterial colony counts. Then, local delivery systems including mucoadhesive, mucopenetrating, and cytoadhesive nanobiomaterials are discussed in the battle against H. pylori infection. Afterward, engineered delivery platforms including polymer-coated nanoemulsions and polymer-coated nanoliposomes are poposed. These bioinspired platforms can contain an antimicrobial agent enclosed within smart multifunctional nanoformulations. These bioplatforms can prevent the development of antibiotic resistance, as well as specifically killing H. pylori with no or only slight negative effects on the host gastrointestinal microbiota. Finally, the essential checkpoints that should be passed to confirm the potential effectiveness of anti-H. pylori nanosystems are discussed.
Collapse
Affiliation(s)
- Mojtaba Kamankesh
- Polymer Chemistry Department, School of Science, University of Tehran, PO Box 14155-6455, Tehran, 14144-6455, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | - Antoni Llopis-Lorente
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Insituto de Salud Carlos III, Valencia, 46022, Spain
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | | | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985717411, Iran
| | - Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, 4 Science Drive 2, Singapore, 117545, Singapore
| |
Collapse
|
13
|
Yu H, Gao R, Liu Y, Fu L, Zhou J, Li L. Stimulus-Responsive Hydrogels as Drug Delivery Systems for Inflammation Targeted Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306152. [PMID: 37985923 PMCID: PMC10767459 DOI: 10.1002/advs.202306152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/19/2023] [Indexed: 11/22/2023]
Abstract
Deregulated inflammations induced by various factors are one of the most common diseases in people's daily life, while severe inflammation can even lead to death. Thus, the efficient treatment of inflammation has always been the hot topic in the research of medicine. In the past decades, as a potential biomaterial, stimuli-responsive hydrogels have been a focus of attention for the inflammation treatment due to their excellent biocompatibility and design flexibility. Recently, thanks to the rapid development of nanotechnology and material science, more and more efforts have been made to develop safer, more personal and more effective hydrogels for the therapy of some frequent but tough inflammations such as sepsis, rheumatoid arthritis, osteoarthritis, periodontitis, and ulcerative colitis. Herein, from recent studies and articles, the conventional and emerging hydrogels in the delivery of anti-inflammatory drugs and the therapy for various inflammations are summarized. And their prospects of clinical translation and future development are also discussed in further detail.
Collapse
Affiliation(s)
- Haoyu Yu
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdong518033P. R. China
| | - Rongyao Gao
- Department of ChemistryRenmin University of ChinaBeijing100872P. R. China
| | - Yuxin Liu
- Department of Biomolecular SystemsMax‐Planck Institute of Colloids and Interfaces14476PotsdamGermany
| | - Limin Fu
- Department of ChemistryRenmin University of ChinaBeijing100872P. R. China
| | - Jing Zhou
- Department of ChemistryCapital Normal UniversityBeijing100048P. R. China
| | - Luoyuan Li
- The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdong518033P. R. China
| |
Collapse
|
14
|
Zou X, Cai S, Wang T, Zheng S, Cui X, Hao J, Chen X, Liu Y, Zhang Z, Li Y. Natural antibacterial agent-based nanoparticles for effective treatment of intracellular MRSA infection. Acta Biomater 2023; 169:410-421. [PMID: 37557944 DOI: 10.1016/j.actbio.2023.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/17/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023]
Abstract
Intracellular MRSA is extremely difficult to eradicate by traditional antibiotics, leading to infection dissemination and drug resistance. A general lack of facile and long-term strategies to effectively eliminate intracellular MRSA. In this study, glabridin (GLA)-loaded pH-responsive nanoparticles (NPs) were constructed using cinnamaldehyde (CA)-dextran conjugates as carriers. These NPs targeted infected macrophages/MRSA via dextran mediation and effectively accumulated at the MRSA infection site. The NPs were then destabilized in response to the low pH of the lysosomes, which triggered the release of CA and GLA. The released CA downregulated the expression of cytotoxic pore-forming toxins, thereby decreasing the damage of macrophage and risk of the intracellular bacterial dissemination. Meanwhile, GLA could rapidly kill intracellularly entrapped MRSA with a low possibility of developing resistance. Using a specific combination of the natural antibacterial agents CA and GLA, NPs effectively eradicated intracellular MRSA with low toxicity to normal tissues in a MRSA-induced peritonitis model. This strategy presents a potential alternative for enhancing intracellular MRSA therapy, particularly for repeated and long-term clinical applications. STATEMENT OF SIGNIFICANCE: Intracellular MRSA infections are a growing threat to public health, and there is a general lack of a facile strategy for efficiently eliminating intracellular MRSA while reducing the ever-increasing drug resistance. In this study, pH-responsive and macrophage/MRSA-targeting nanoparticles were prepared by conjugating the phytochemical cinnamaldehyde to dextran to encapsulate the natural antibacterial agent glabridin. Using a combination of traditional Chinese medicine, the NPs significantly increased drug accumulation in MRSA and showed superior intracellular and extracellular bactericidal activity. Importantly, the NPs can inhibit potential intracellular bacteria dissemination and reduce the development of drug resistance, thus allowing for repeated treatment. Natural antibacterial agent-based drug delivery systems are an attractive alternative for facilitating the clinical treatment of intracellular MRSA infections.
Collapse
Affiliation(s)
- Xinshu Zou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Shuang Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Tingting Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Sidi Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Xilong Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Jingyou Hao
- Harbin Lvdasheng Animal Medicine Manufacture Co., Ltd., Harbin 150000, PR China
| | - Xueying Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Yanyan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China
| | - Zhiyun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China.
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, PR China.
| |
Collapse
|
15
|
Duan QY, Zhu YX, Jia HR, Wang SH, Wu FG. Nanogels: Synthesis, properties, and recent biomedical applications. PROGRESS IN MATERIALS SCIENCE 2023; 139:101167. [DOI: 10.1016/j.pmatsci.2023.101167] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Karmakar S, Shanmugasundaram S, Modak B. Oleogel-based drug delivery for the treatment of periodontitis: current strategies and future perspectives. F1000Res 2023; 12:1228. [PMID: 38690138 PMCID: PMC11058454 DOI: 10.12688/f1000research.140173.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2023] [Indexed: 05/02/2024] Open
Abstract
Periodontitis is the chronic inflammation of tooth-supporting tissues that leads to loss of tooth support if untreated. Conventional therapy for periodontitis (mechanical removal of microbial biofilm and oral hygiene enforcement) is augmented by anti-microbial and anti-inflammatory drugs. These drugs are frequently delivered locally into the periodontal pocket for maximum efficiency and minimum adverse effects. The potential of oleogels for periodontal drug delivery has been discussed and further, the future scope of oleogel-based drug delivery systems in dentistry. An oleogel-based local drug delivery system offers several advantages over other systems. Superior mechanical properties (firmness and compressibility), muco-adhesion, shear thinning, thixotropy, controlled drug release and the ability to incorporate water-insoluble drugs clearly distinguish and highlight the potential of oleogels as periodontal local drug delivery systems. Bigels can combine the qualities of both hydrogels and oleogels to provide a more promising option for drug delivery. However, there is limited evidence concerning oleogels as local drug delivery agents in periodontics. Further studies are needed to discern the clinical efficacy of oleogel-based drug delivery systems.
Collapse
Affiliation(s)
- Shaswata Karmakar
- Department of Periodontology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shashikiran Shanmugasundaram
- Department of Periodontology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Baishakhi Modak
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
17
|
Hadiya S, Ibrahem RA, Abd El-Baky RM, Elsabahy M, Hussein AM, Tolba ME, Aly SA. Nano-ciprofloxacin/meropenem exhibit bactericidal activity against Gram-negative bacteria and rescue septic rat model. Nanomedicine (Lond) 2023; 18:1553-1566. [PMID: 37933674 DOI: 10.2217/nnm-2022-0314] [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] [Indexed: 11/08/2023] Open
Abstract
Aim: We hypothesized that simultaneous administration of two antibiotics loaded into a nanopolymer matrix would augment their synergistic bactericidal interaction. Methods: Nanoplatforms of chitosan/Pluronic® loaded with ciprofloxacin/meropenem (CS/Plu-Cip/Mer) were prepared by the ionic gelation method, using Plu at concentrations in the range 0.5-4% w/v. CS/Plu-Cip/Mer was evaluated for antibacterial synergistic activity in vitro and in vivo. Results: CS/Plu-Cip and CS/Plu-Mer with Plu concentrations of 3% w/v and 2% w/v, respectively, exhibited ∼80% encapsulation efficiency. The MICs of pathogens were fourfold to 16-fold lower for CS/Plu-Cip/Mer than for Cip/Mer. Synergy was evidenced for CS/Plu-Cip/Mer with a bactericidal effect (at 1× MIC and sub-MICs), and it significantly decreased bacterial load and rescued infected rats. Conclusion: This study illustrates the ability of CS/Plu nanopolymer to intensify synergy between antibiotics, thereby providing a promising potential to rejuvenate antibiotics considered ineffective against resistant pathogens.
Collapse
Affiliation(s)
- Safy Hadiya
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, 71515, Egypt
| | - Reham A Ibrahem
- Department of Microbiology & Immunology, Faculty of Pharmacy, Minia University, Minia, 61511, Egypt
| | - Rehab M Abd El-Baky
- Department of Microbiology & Immunology, Faculty of Pharmacy, Minia University, Minia, 61511, Egypt
- Department of Microbiology & Immunology, Faculty of Pharmacy, Deraya University, Minia, 61511, Egypt
| | - Mahmoud Elsabahy
- School of Biotechnology, Badr University in Cairo, Badr City, 11829, Egypt
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Abeer Mr Hussein
- Pharmacology Department, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Mohammed Em Tolba
- Medical Parasitology Department, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Sherine A Aly
- Department of Microbiology & Immunology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| |
Collapse
|
18
|
Kesharwani P, Prajapati SK, Jain A, Sharma S, Mody N, Jain A. Biodegradable Nanogels for Dermal Applications: An Insight. CURRENT NANOSCIENCE 2023; 19:509-524. [DOI: 10.2174/1573413718666220415095630] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/12/2021] [Accepted: 12/23/2021] [Indexed: 08/22/2024]
Abstract
Abstract:
Biodegradable nanogels in the biomedical field are emerging vehicles comprising
dispersions of hydrogel nanoparticles having 3D crosslinked polymeric networks. Nanogels show
distinguished characteristics including their homogeneity, adjustable size, low toxicity, stability
in serum, stimuli-responsiveness (pH, temperature, enzymes, light, etc.), and relatively good
drug encapsulation capability. Due to these characteristics, nanogels are referred to as nextgeneration
drug delivery systems and are suggested as promising carriers for dermal applications.
The site-specific delivery of drugs with effective therapeutic effects is crucial in transdermal drug
delivery. The nanogels made from biodegradable polymers can show external stimuliresponsiveness
which results in a change in gel volume, water content, colloidal stability, mechanical
strength, and other physical and chemical properties, thus improving the site-specific
topical drug delivery. This review provides insight into the advances in development, limitations,
and therapeutic significance of nanogels formulations. It also highlights the process of release of
drugs in response to external stimuli, various biodegradable polymers in the formulation of the
nanogels, and dermal applications of nanogels and their role in imaging, anti‐inflammatory therapy,
antifungal and antimicrobial therapy, anti‐psoriatic therapy, and ocular and protein/peptide
drug delivery.
Collapse
Affiliation(s)
- Payal Kesharwani
- Institute of Pharmacy, Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, 201310, Uttar Pradesh,
India
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O. Rajasthan 304022, India
| | - Shiv Kumar Prajapati
- Institute of Pharmacy, Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, 201310, Uttar Pradesh,
India
| | - Anushka Jain
- Raj Kumar
Goel Institute of Technology (Pharmacy), 5-Km. Stone, Delhi-Meerut Road, Ghaziabad, Uttar Pradesh, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O. Rajasthan-304022-India
| | - Nishi Mody
- Department of Pharmaceutical Sciences, Dr. H. S. Gour University, Sagar (MP) 470003, India
| | - Ankit Jain
- Department of
Materials Engineering, Indian Institute of Science, Bangalore 560012 (Karnataka), India
| |
Collapse
|
19
|
Luo W, Meng K, Zhao Y, Liu J, Chen D, Xu C, Algharib SA, Dawood AS, Xie S. Guar gum modified tilmicosin-loaded sodium alginate/gelatin composite nanogels for effective therapy of porcine proliferative enteritis caused by Lawsonia intracellularis. Int J Biol Macromol 2023:125084. [PMID: 37245769 DOI: 10.1016/j.ijbiomac.2023.125084] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 05/30/2023]
Abstract
In order to overcome the treatment difficulty of Lawsonia intracellularis (L.intracellularis) using antibiotics, the tilmicosin (TIL)-loaded sodium alginate (SA)/gelatin composite nanogels modified with bioadhesive substances were designed. The optimized nanogels were prepared by electrostatic interaction between SA and gelatin at a mass ratio of 1:1 and CaCl2 as an ionic crosslinker and further modified with guar gum (GG). The optimized TIL-nanogels modified with GG had a uniform spherical shape with a diameter of 18.2 ± 0.3 nm, LC of 29.4 ± 0.2 %, EE of 70.4 ± 1.6 %, PDI of 0.30 ± 0.04, and ZP of -32.2 ± 0.5 mv. The FTIR, DSC, and PXRD showed that GG was covered on the surface of TIL-nanogels in a pattern of staggered arrangements. The TIL-nanogels modified with GG had the strongest adhesive strength amongst those with I-carrageenan and locust bean gum and the plain nanogels, and thus significantly enhanced the cellular uptake and accumulation of TIL via clathrin-mediated endocytosis. It exhibited an increased therapeutic effect against L.intracellularis in vitro and in vivo. This study will provide guidance for developing nanogels for intracellular bacterial infection treatment.
Collapse
Affiliation(s)
- Wanhe Luo
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Engineering Laboratory for Tarim Animal Diseases Diagnosis and Control, College of Animal Science and Technology, Tarim University, Alar, Xinjiang 843300, China
| | - Kuiyu Meng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yiqing Zhao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jinhuan Liu
- Engineering Laboratory for Tarim Animal Diseases Diagnosis and Control, College of Animal Science and Technology, Tarim University, Alar, Xinjiang 843300, China
| | - Dongmei Chen
- MARA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Chunyan Xu
- Engineering Laboratory for Tarim Animal Diseases Diagnosis and Control, College of Animal Science and Technology, Tarim University, Alar, Xinjiang 843300, China
| | - Samah Attia Algharib
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, QG, Egypt
| | - Ali Sobhy Dawood
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Medicine and Infectious Diseases Department, Faculty of Veterinary Medicine, University of Sadat City, 32897, Egypt
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
20
|
Hemmingsen LM, Panzacchi V, Kangu LM, Giordani B, Luppi B, Škalko-Basnet N. Lecithin and Chitosan as Building Blocks in Anti- Candida Clotrimazole Nanoparticles. Pharmaceuticals (Basel) 2023; 16:790. [PMID: 37375738 DOI: 10.3390/ph16060790] [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: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
The main focus when considering treatment of non-healing and infected wounds is tied to the microbial, particularly bacterial, burden within the wound bed. However, as fungal contributions in these microbial communities become more recognized, the focus needs to be broadened, and the remaining participants in the complex wound microbiome need to be addressed in the development of new treatment strategies. In this study, lecithin/chitosan nanoparticles loaded with clotrimazole were tailored to eradicate one of the most abundant fungi in the wound environment, namely C. albicans. Moreover, this investigation was extended to the building blocks and their organization within the delivery system. In the evaluation of the novel nanoparticles, their compatibility with keratinocytes was confirmed. Furthermore, these biocompatible, biodegradable, and non-toxic carriers comprising clotrimazole (~189 nm, 24 mV) were evaluated for their antifungal activity through both disk diffusion and microdilution methods. It was found that the activity of clotrimazole was fully preserved upon its incorporation into this smart delivery system. These results indicate both that the novel carriers for clotrimazole could serve as a therapeutic alternative in the treatment of fungi-infected wounds and that the building blocks and their organization affect the performance of nanoparticles.
Collapse
Affiliation(s)
- Lisa Myrseth Hemmingsen
- Department of Pharmacy, University of Tromsø-The Arctic University of Norway, Universitetsvegen 57, 9037 Tromsø, Norway
| | - Virginia Panzacchi
- Department of Pharmacy, University of Tromsø-The Arctic University of Norway, Universitetsvegen 57, 9037 Tromsø, Norway
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy
| | - Lloyd Mbugua Kangu
- Department of Pharmacy, University of Tromsø-The Arctic University of Norway, Universitetsvegen 57, 9037 Tromsø, Norway
| | - Barbara Giordani
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy
| | - Barbara Luppi
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127 Bologna, Italy
| | - Nataša Škalko-Basnet
- Department of Pharmacy, University of Tromsø-The Arctic University of Norway, Universitetsvegen 57, 9037 Tromsø, Norway
| |
Collapse
|
21
|
Ye Y, Yu H, Chen B, Zhao Y, Lv B, Xue G, Sun Y, Cao J. Engineering nanoenzymes integrating Iron-based metal organic frameworks with Pt nanoparticles for enhanced Photodynamic-Ferroptosis therapy. J Colloid Interface Sci 2023; 645:882-894. [PMID: 37178565 DOI: 10.1016/j.jcis.2023.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Photodynamic therapy (PDT), as a promising strategy in cancer treatment that utilizes photosensitizers (PSs) to produce reactive oxygen species, has been widely used for eliminating cancer cells under specific wavelength light irradiation. However, the low aqueous solubility of PSs and special tumor microenvironments (TME), such as high glutathione (GSH) and tumor hypoxia remain challenges towards PDT for hypoxic tumor treatment. To address these problems, we constructed a novel nanoenzyme for enhanced PDT-ferroptosis therapy by integrating small Pt nanoparticles (Pt NPs) and near-infrared photosensitizer CyI into iron-based metal organic frameworks (MOFs). In addition, hyaluronic acid was adhered to the surface of the nanoenzymes to enhance the targeting ability. In this design, MOFs act not only as a delivery vector for PSs, but also a ferroptosis inducer. Pt NPs stabilized by MOFs were functioned as an oxygen (O2) generator by catalyzing hydrogen peroxide into O2 to relieve tumor hypoxia and increase singlet oxygen generation. In vitro and in vivo results demonstrated that under laser irradiation, this nanoenzyme could effectively relive the tumor hypoxia and decrease the level of GSH, resulting in enhanced PDT-ferroptosis therapy against hypoxic tumor. The proposed nanoenzymes represent an important advance in altering TME for improved clinical PDT-ferroptosis therapy, as well as their potential as effective theranostic agents for hypoxic tumors.
Collapse
Affiliation(s)
- Yuyun Ye
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Hongli Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Bohan Chen
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Yifan Zhao
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Bai Lv
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Guanghe Xue
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Jie Cao
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
22
|
Zhao D, Feng W, Kang X, Li H, Liu F, Zheng W, Li G, Wang X. Dual-targeted poly(amino acid) nanoparticles deliver drug combinations on-site: an intracellular synergistic strategy to eliminate intracellular bacteria. J Mater Chem B 2023; 11:2958-2971. [PMID: 36919349 DOI: 10.1039/d3tb00125c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Multi-drug combinations are a common strategy for the treatment of intracellular bacterial infections. However, different internalized pathways and the accumulation of the composite drugs at different subcellular organelles very much reduce their efficacy. Herein, an intracellular synergistic strategy is proposed, which is realized by on-site delivery of a drug combination using a macrophage/intracellular bacterium-dual targeted drug delivery system (DDS). The DDS is fabricated by encapsulating vancomycin (Van) and curcumin (Cur) into poly(α-N-acryloyl-phenylalanine)-block-poly(β-N-acryloyl-D-aminoalanine-co-2-O-acetyl-α-D-mannosyloxy) nanoparticles, denoted by (Van + Cur)@F(AM) NPs. Mannose ligands on (Van + Cur)@F(AM) NPs trigger their specific internalization in macrophages, while aminoalanine moieties subsequently drive the NPs to target intracellular methicillin-resistant Staphylococcus aureus (MRSA). Thereafter, Van and Cur are durably released in a synergistic dose at the residence site of intracellular MRSA. Under this intracellular synergistic effect, (Van + Cur)@F(AM) NPs show superior elimination efficiency in vitro and in vivo compared to the control groups, including free Van, (Van + Cur), the DDS encapsulated Van and the DDSs separately-encapsulated Van and Cur. Furthermore, (Van + Cur)@F(AM) NPs significantly enhance the in vivo antibacterial capacity by modulating the immune response. Therefore, this dual-targeted DDS-assisted intracellular synergistic antibacterial strategy of drug combination is an effective therapeutic against intracellular bacteria.
Collapse
Affiliation(s)
- Dongdong Zhao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Xiaoxu Kang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Haofei Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Fang Liu
- Department of Oncology of Integrative Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Weitao Zheng
- Hubei Provincial Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, Hubei Province, China
| | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| |
Collapse
|
23
|
Qu S, Zhu K. Endocytosis-mediated redistribution of antibiotics targets intracellular bacteria. NANOSCALE 2023; 15:4781-4794. [PMID: 36779877 DOI: 10.1039/d2nr05421c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The increasing emergence and dissemination of antibiotic resistance pose a severe threat to overwhelming healthcare practices worldwide. The lack of new antibacterial drugs urgently calls for alternative therapeutic strategies to combat multidrug-resistant (MDR) bacterial pathogens, especially those that survive and replicate in host cells, causing relapse and recurrence of infections. Intracellular drug delivery is a direct efficient strategy to combat invasive pathogens by increasing the accumulation of antibiotics. However, the increased accumulation of antibiotics in the infected host cells does not mean high efficacy. The difficulty of treatment lies in the efficient intracellular delivery of antibiotics to the pathogen-containing compartments. Here, we first briefly review the survival mechanisms of intracellular bacteria to facilitate the exploration of potential antibacterial targets for precise delivery. Furthermore, we provide an overview of endocytosis-mediated drug delivery systems, including the biomedical and physicochemical properties modulating the endocytosis and intracellular redistribution of antibiotics. Lastly, we summarize the targets and payloads of recently described intracellular delivery systems and their modes of action against diverse pathogenic bacteria-associated infections. This overview of endocytosis-mediated redistribution of antibiotics sheds light on the development of novel delivery platforms and alternative strategies to combat intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Kui Zhu
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China.
- Engineering Research Center of Animal Innovative drugs and Safety Evaluation, Ministry of Education, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
24
|
Feng W, Chittò M, Moriarty TF, Li G, Wang X. Targeted Drug Delivery Systems for Eliminating Intracellular Bacteria. Macromol Biosci 2023; 23:e2200311. [PMID: 36189899 DOI: 10.1002/mabi.202200311] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Indexed: 01/19/2023]
Abstract
The intracellular survival of pathogenic bacteria requires a range of survival strategies and virulence factors. These infections are a significant clinical challenge, wherein treatment frequently fails because of poor antibiotic penetration, stability, and retention in host cells. Drug delivery systems (DDSs) are promising tools to overcome these shortcomings and enhance the efficacy of antibiotic therapy. In this review, the classification and the mechanisms of intracellular bacterial persistence are elaborated. Furthermore, the systematic design strategies applied to DDSs to eliminate intracellular bacteria are also described, and the strategies used for internalization, intracellular activation, bacterial targeting, and immune enhancement are highlighted. Finally, this overview provides guidance for constructing functionalized DDSs to effectively eliminate intracellular bacteria.
Collapse
Affiliation(s)
- Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,AO Research Institute Davos, Davos, 7270, Switzerland
| | - Marco Chittò
- AO Research Institute Davos, Davos, 7270, Switzerland
| | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
25
|
Gbian DL, Omri A. Lipid-Based Drug Delivery Systems for Diseases Managements. Biomedicines 2022; 10:2137. [PMID: 36140237 PMCID: PMC9495957 DOI: 10.3390/biomedicines10092137] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022] Open
Abstract
Liposomes are tiny lipid-based vesicles composed of one or more lipid bilayers, which facilitate the encapsulation of hydrophilic, lipophilic, and amphiphilic biological active agents. The description of the physicochemical properties, formulation methods, characteristics, mechanisms of action, and large-scale manufacturing of liposomes as delivery systems are deeply discussed. The benefits, toxicity, and limitations of the use of liposomes in pharmacotherapeutics including in diagnostics, brain targeting, eye and cancer diseases, and in infections are provided. The experimental approaches that may reduce, or even bypass, the use of liposomal drug drawbacks is described. The application of liposomes in the treatment of numerous diseases is discussed.
Collapse
Affiliation(s)
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
26
|
Hanke M, Grundmeier G, Keller A. Direct visualization of the drug loading of single DNA origami nanostructures by AFM-IR nanospectroscopy. NANOSCALE 2022; 14:11552-11560. [PMID: 35861612 DOI: 10.1039/d2nr02701a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The efficient loading of DNA nanostructures with intercalating or groove-binding drugs is an important prerequisite for various applications in drug delivery. However, unambiguous verification and quantification of successful drug loading is often rather challenging. In this work, AFM-IR nanospectroscopy is thus employed to directly visualize the loading of DNA origami nanostructures with the photosensitizer methylene blue (MB). Single MB-loaded DNA origami nanostructures can be clearly resolved in high-resolution infrared (IR) maps and the occurrence of MB-specific IR absorption correlates well with the topographic signals of the DNA origami nanostructures. The intensity of the recorded MB absorption bands furthermore scales with the MB concentration used for MB loading. By comparing single- and multilayer DNA origami nanostructures, it is also shown that the IR signal intensity of the loaded MB increases with the thickness of the DNA origami nanostructures. This indicates that also DNA double helices located in the core of bulky 3D DNA origami nanostructures are accessible for MB loading. AFM-IR nanospectroscopy thus has the potential to become an invaluable tool for quantifying drug loading of DNA origami nanostructures and optimizing drug loading protocols.
Collapse
Affiliation(s)
- Marcel Hanke
- Paderborn University, Technical and Macromolecular Chemistry, Warburger Str. 100, 33098 Paderborn, Germany.
| | - Guido Grundmeier
- Paderborn University, Technical and Macromolecular Chemistry, Warburger Str. 100, 33098 Paderborn, Germany.
| | - Adrian Keller
- Paderborn University, Technical and Macromolecular Chemistry, Warburger Str. 100, 33098 Paderborn, Germany.
| |
Collapse
|
27
|
Lukhey MS, Shende P. Advancement in wound healing treatment using functional nanocarriers. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2099393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Mihir S. Lukhey
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Mumbai, India
| |
Collapse
|
28
|
Zong TX, Silveira AP, Morais JAV, Sampaio MC, Muehlmann LA, Zhang J, Jiang CS, Liu SK. Recent Advances in Antimicrobial Nano-Drug Delivery Systems. NANOMATERIALS 2022; 12:nano12111855. [PMID: 35683711 PMCID: PMC9182179 DOI: 10.3390/nano12111855] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/28/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022]
Abstract
Infectious diseases are among the major health issues of the 21st century. The substantial use of antibiotics over the years has contributed to the dissemination of multidrug resistant bacteria. According to a recent report by the World Health Organization, antibacterial (ATB) drug resistance has been one of the biggest challenges, as well as the development of effective long-term ATBs. Since pathogens quickly adapt and evolve through several strategies, regular ATBs usually may result in temporary or noneffective treatments. Therefore, the demand for new therapies methods, such as nano-drug delivery systems (NDDS), has aroused huge interest due to its potentialities to improve the drug bioavailability and targeting efficiency, including liposomes, nanoemulsions, solid lipid nanoparticles, polymeric nanoparticles, metal nanoparticles, and others. Given the relevance of this subject, this review aims to summarize the progress of recent research in antibacterial therapeutic drugs supported by nanobiotechnological tools.
Collapse
Affiliation(s)
- Tong-Xin Zong
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (T.-X.Z.); (J.Z.)
| | - Ariane Pandolfo Silveira
- Institute of Biological Sciences, University of Brasília, Brasilia 70910900, Brazil; (A.P.S.); (J.A.V.M.); (M.C.S.)
| | | | - Marina Carvalho Sampaio
- Institute of Biological Sciences, University of Brasília, Brasilia 70910900, Brazil; (A.P.S.); (J.A.V.M.); (M.C.S.)
| | - Luis Alexandre Muehlmann
- Institute of Biological Sciences, University of Brasília, Brasilia 70910900, Brazil; (A.P.S.); (J.A.V.M.); (M.C.S.)
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220900, Brazil
- Correspondence: (L.A.M.); (C.-S.J.); (S.-K.L.)
| | - Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (T.-X.Z.); (J.Z.)
| | - Cheng-Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (T.-X.Z.); (J.Z.)
- Correspondence: (L.A.M.); (C.-S.J.); (S.-K.L.)
| | - Shan-Kui Liu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; (T.-X.Z.); (J.Z.)
- Correspondence: (L.A.M.); (C.-S.J.); (S.-K.L.)
| |
Collapse
|
29
|
Padmakumar A, Koyande NP, Rengan AK. The Role of Hitchhiking in Cancer Therapeutics – A review. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Ananya Padmakumar
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad Sangareddy 502284 India
| | - Navami Prabhakar Koyande
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad Sangareddy 502284 India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad Sangareddy 502284 India
| |
Collapse
|
30
|
Jebastin K, Narayanasamy D. Rationale utilization of phospholipid excipients: a distinctive tool for progressing state of the art in research of emerging drug carriers. J Liposome Res 2022; 33:1-33. [PMID: 35543241 DOI: 10.1080/08982104.2022.2069809] [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: 10/18/2022]
Abstract
Phospholipids have a high degree of biocompatibility and are deemed ideal pharmaceutical excipients in the development of lipid-based drug delivery systems, because of their unique features (permeation, solubility enhancer, emulsion stabilizer, micelle forming agent, and the key excipients in solid dispersions) they can be used in a variety of pharmaceutical drug delivery systems, such as liposomes, phytosomes, solid lipid nanoparticles, etc. The primary usage of phospholipids in a colloidal pharmaceutical formulation is to enhance the drug's bioavailability with low aqueous solubility [i.e. Biopharmaceutical Classification System (BCS) Class II drugs], Membrane penetration (i.e. BCS Class III drugs), drug uptake and release enhancement or modification, protection of sensitive active pharmaceutical ingredients (APIs) from gastrointestinal degradation, a decrease of gastrointestinal adverse effects, and even masking of the bitter taste of orally delivered drugs are other uses. Phospholipid-based colloidal drug products can be tailored to address a wide variety of product requirements, including administration methods, cost, product stability, toxicity, and efficacy. Such formulations that are also a cost-effective method for developing medications for topical, oral, pulmonary, or parenteral administration. The originality of this review work is that we comprehensively evaluated the unique properties and special aspects of phospholipids and summarized how the individual phospholipids can be utilized in various types of lipid-based drug delivery systems, as well as listing newly marketed lipid-based products, patents, and continuing clinical trials of phospholipid-based therapeutic products. This review would be helpful for researchers responsible for formulation development and research into novel colloidal phospholipid-based drug delivery systems.
Collapse
Affiliation(s)
- Koilpillai Jebastin
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, India
| | - Damodharan Narayanasamy
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
31
|
Obuobi S, Ngoc Phung A, Julin K, Johannessen M, Škalko-Basnet N. Biofilm Responsive Zwitterionic Antimicrobial Nanoparticles to Treat Cutaneous Infection. Biomacromolecules 2021; 23:303-315. [PMID: 34914360 PMCID: PMC8753600 DOI: 10.1021/acs.biomac.1c01274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
To avert the poor
bioavailability of antibiotics during S. aureus biofilm
infections, a series of zwitterionic nanoparticles
containing nucleic acid nanostructures were fabricated for the delivery
of vancomycin. The nanoparticles were prepared with three main lipids:
(i) neutral (soy phosphatidylcholine; P), (ii) positively charged
ionizable (1,2-dioleyloxy-3-dimethylaminopropane; D), and (iii) anionic
(1,2-dipalmitoyl-sn-glycero-3-phospho((ethyl-1′,2′,3′-triazole)
triethylene glycolmannose; M) or (cholesteryl hemisuccinate; C) lipids.
The ratio of the anionic lipid was tuned between 0 and 10 mol %, and
its impact on surface charge, size, stability, toxicity, and biofilm
sensitivity was evaluated. Under biofilm mimicking conditions, the
enzyme degradability (via dynamic light scattering (DLS)), antitoxin
(via DLS and spectrophotometry), and antibiotic release profile was
assessed. Additionally, biofilm penetration, prevention (in
vitro), and eradication (ex vivo) of the
vancomycin loaded formulation was investigated. Compared with the
unmodified nanoparticles which exhibited the smallest size (188 nm),
all three surface modified formulations showed significantly larger
sizes (i.e., 222–277 nm). Under simulations of biofilm pH conditions,
the mannose modified nanoparticle (PDM 90/5/5) displayed ideal charge
reversal from a neutral (+1.69 ± 1.83 mV) to a cationic surface
potential (+17.18 ± 2.16 mV) to improve bacteria binding and
biofilm penetration. In the presence of relevant bacterial enzymes,
the carrier rapidly released the DNA nanoparticles to function as
an antitoxin against α-hemolysin. Controlled release of vancomycin
prevented biofilm attachment and significantly reduced early stage
biofilm formations within 24 h. Enhanced biocompatibility and significant ex vivo potency of the PDM 90/5/5 formulation was also observed.
Taken together, these results emphasize the benefit of these nanocarriers
as potential therapies against biofilm infections and fills the gap
for multifunctional nanocarriers that prevent biofilm infections.
Collapse
Affiliation(s)
- Sybil Obuobi
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø 9037, Norway
| | - Anna Ngoc Phung
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø 9037, Norway
| | - Kjersti Julin
- Host Microbe Interaction research group, Department of Medical Biology, UIT The Arctic University of Norway, Tromsø 9037, Norway
| | - Mona Johannessen
- Host Microbe Interaction research group, Department of Medical Biology, UIT The Arctic University of Norway, Tromsø 9037, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø 9037, Norway
| |
Collapse
|
32
|
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: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [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
|
33
|
Development of a Polysaccharide-Based Hydrogel Drug Delivery System (DDS): An Update. Gels 2021; 7:gels7040153. [PMID: 34698125 PMCID: PMC8544468 DOI: 10.3390/gels7040153] [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: 07/29/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Delivering a drug to the target site with minimal-to-no off-target cytotoxicity is the major determinant for the success of disease therapy. While the therapeutic efficacy and cytotoxicity of the drug play the main roles, the use of a suitable drug delivery system (DDS) is important to protect the drug along the administration route and release it at the desired target site. Polysaccharides have been extensively studied as a biomaterial for DDS development due to their high biocompatibility. More usefully, polysaccharides can be crosslinked with various molecules such as micro/nanoparticles and hydrogels to form a modified DDS. According to IUPAC, hydrogel is defined as the structure and processing of sols, gels, networks and inorganic–organic hybrids. This 3D network which often consists of a hydrophilic polymer can drastically improve the physical and chemical properties of DDS to increase the biodegradability and bioavailability of the carrier drugs. The advancement of nanotechnology also allows the construction of hydrogel DDS with enhanced functionalities such as stimuli-responsiveness, target specificity, sustained drug release, and therapeutic efficacy. This review provides a current update on the use of hydrogel DDS derived from polysaccharide-based materials in delivering various therapeutic molecules and drugs. We also highlighted the factors that affect the efficacy of these DDS and the current challenges of developing them for clinical use.
Collapse
|
34
|
Bhattacharyya S, Sudheer P, Das K, Ray S. Experimental Design Supported Liposomal Aztreonam Delivery: In Vitro Studies. Adv Pharm Bull 2021; 11:651-662. [PMID: 34888212 PMCID: PMC8642795 DOI: 10.34172/apb.2021.074] [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: 08/25/2020] [Accepted: 09/15/2020] [Indexed: 11/09/2022] Open
Abstract
Purpose: The present study focuses on a systemic approach to develop liposomal aztreonam as a promising dosage form for inhalation therapy in the treatment of pneumonia and explores the in-vitro antimicrobial and cell uptake efficacy. Methods: Liposomes were prepared by ethanol injection method using the lipids - soya phosphatidylcholine (SP) and cholesterol (CH). A central composite design (CCD) was employed to optimize the lipid composition to evaluate the effect on vesicle size, zeta potential and entrapment efficiency of the formulation. A numerical and graphical optimization was carried out to predict the optimized blend. The optimized formulation was characterized for vesicle size, surface charge, encapsulation, surface morphology, differential scanning calorimetry (DSC), powder X Ray Diffraction (PXRD), thermogravimetric analysis (TGA), in vitro diffusion, accelerated stability studies, antimicrobial studies on Pseudomonas aeruginosa NCIM 2200 and in vitro cell uptake studies. Results: The optimized formulation was found to have a particle size of 144 nm, a surface charge of -35 mV, with satisfactory drug entrapment. The surface morphology study proved the formation of nanosized vesicles. The drug release from liposomal matrix was biphasic in nature. The solid-state study revealed the reason for good encapsulation of drug. The moisture retention capacity was found to be minimum. The anti-microbial study revealed the potential antibacterial activity of the optimized formulation over the pure drug. The formulation was found to be safe on the epithelial cells and showed a marked increase in cellular uptake of aztreonam in a lipid carrier. Conclusion: It can be concluded that the optimized liposomal aztreonam could be considered as a promising approach for the delivery of aztreonam through inhalation.
Collapse
Affiliation(s)
| | - Preethi Sudheer
- Krupanidhi College of Pharmacy, Bengaluru, Karnataka 560035, India
| | - Kuntal Das
- Krupanidhi College of Pharmacy, Bengaluru, Karnataka 560035, India
| | - Subhabrata Ray
- Dr. BC Roy College of Pharmacy, Durgapur, West Bengal 713206, India
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Gbian DL, Omri A. The Impact of an Efflux Pump Inhibitor on the Activity of Free and Liposomal Antibiotics against Pseudomonas aeruginosa. Pharmaceutics 2021; 13:pharmaceutics13040577. [PMID: 33919624 PMCID: PMC8072581 DOI: 10.3390/pharmaceutics13040577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 11/21/2022] Open
Abstract
The eradication of Pseudomonas aeruginosa in cystic fibrosis patients has become continuously difficult due to its increased resistance to treatments. This study assessed the efficacy of free and liposomal gentamicin and erythromycin, combined with Phenylalanine arginine beta-naphthylamide (PABN), a broad-spectrum efflux pump inhibitor, against P. aeruginosa isolates. Liposomes were prepared and characterized for their sizes and encapsulation efficiencies. The antimicrobial activities of formulations were determined by the microbroth dilution method. Their activity on P. aeruginosa biofilms was assessed, and the effect of sub-inhibitory concentrations on bacterial virulence factors, quorum sensing (QS) signals and bacterial motility was also evaluated. The average diameters of liposomes were 562.67 ± 33.74 nm for gentamicin and 3086.35 ± 553.95 nm for erythromycin, with encapsulation efficiencies of 13.89 ± 1.54% and 51.58 ± 2.84%, respectively. Liposomes and PABN combinations potentiated antibiotics by reducing minimum inhibitory and bactericidal concentrations by 4–32 fold overall. The formulations significantly inhibited biofilm formation and differentially attenuated virulence factor production as well as motility. Unexpectedly, QS signal production was not affected by treatments. Taken together, the results indicate that PABN shows potential as an adjuvant of liposomal macrolides and aminoglycosides in the management of lung infections in cystic fibrosis patients.
Collapse
Affiliation(s)
| | - Abdelwahab Omri
- Correspondence: ; Tel.: +1-705-675-1151-2190; Fax: +1-705-675-4844
| |
Collapse
|
38
|
Antimicrobial nanomedicine for ocular bacterial and fungal infection. Drug Deliv Transl Res 2021; 11:1352-1375. [PMID: 33840082 DOI: 10.1007/s13346-021-00966-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
Ocular infection induced by bacteria and fungi is a major cause of visual impairment and blindness. Topical administration of antibiotics remains the first-line treatment, as effective eradication of pathogens is the core of the anti-infection strategy. Whereas, eye drops lack efficiency and have relatively low bioavailability. Intraocular injection may cause concurrent ocular damage and secondary infection. In addition, antibiotic-based management can be limited by the low sensitivity to multidrug-resistant bacteria. Nanomedicine is proposed as a prospective, effective, and noninvasive platform to mediate ocular delivery and combat pathogen or even resistant strains. Nanomedicine can not only carry antimicrobial agents to fight against pathogens but also directly active microbicidal capability, killing pathogens. More importantly, by modification, nanomedicine can achieve enhanced residence time and release time on the cornea, and easy penetration through corneal tissues into anterior and posterior segments of the eye, thus improving the therapeutic effect for ocular infection. In this review, several categories of antimicrobial nanomedicine are systematically discussed, where the efficiency and possibility of further embellishment and improvement to adapt to clinical use are also investigated. All in all, novel antimicrobial nanomedicine provides potent and prospective ways to manage severe and refractory ocular infections.
Collapse
|
39
|
Nanogels Capable of Triggered Release. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2021; 178:99-146. [PMID: 33665715 DOI: 10.1007/10_2021_163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
This chapter provides an overview of soft and environmentally sensitive polymeric nanosystems, which are widely known as nanogels. These particles keep great promise to the area of drug delivery due to their high biocompatibility with body fluids and tissues, as well as due to their ability to encapsulate and release the loaded drugs in a controlled manner. For a long period of time, the controlled drug delivery systems were designed to provide long-termed or sustained release. However, some medical treatments such as cancer chemotherapy, protein and gene delivery do not require the prolonged release of the drug in the site of action. In contrast, the rapid increase of the drug concentration is needed for gaining the desired biological effect. Being very sensitive to surrounding media and different stimuli, nanogels can undergo physico-chemical transitions or chemical changes in their structure. Such changes can result in more rapid release of the drugs, which is usually referred to as triggered drug release. Herein we give the basic information on nanogel unique features, methods of sensitive nanogels preparation, as well as on main mechanisms of triggered release. Additionally, the triggered release of low-molecular drugs and biomacromolecules are discussed.
Collapse
|
40
|
Smith DM, Keller A. DNA Nanostructures in the Fight Against Infectious Diseases. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000049. [PMID: 33615315 PMCID: PMC7883073 DOI: 10.1002/anbr.202000049] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Throughout history, humanity has been threatened by countless epidemic and pandemic outbreaks of infectious diseases, from the Justinianic Plague to the Spanish flu to COVID-19. While numerous antimicrobial and antiviral drugs have been developed over the last 200 years to face these threats, the globalized and highly connected world of the 21st century demands for an ever-increasing efficiency in the detection and treatment of infectious diseases. Consequently, the rapidly evolving field of nanomedicine has taken up the challenge and developed a plethora of strategies to fight infectious diseases with the help of various nanomaterials such as noble metal nanoparticles, liposomes, nanogels, and virus capsids. DNA nanotechnology represents a comparatively recent addition to the nanomedicine arsenal, which, over the past decade, has made great progress in the area of cancer diagnostics and therapy. However, the past few years have seen also an increasing number of DNA nanotechnology-related studies that particularly focus on the detection and inhibition of microbial and viral pathogens. Herein, a brief overview of this rather young research field is provided, successful concepts as well as potential challenges are identified, and promising directions for future research are highlighted.
Collapse
Affiliation(s)
- David M. Smith
- DNA Nanodevices UnitDepartment DiagnosticsFraunhofer Institute for Cell Therapy and Immunology IZI04103LeipzigGermany
- Peter Debye Institute for Soft Matter PhysicsFaculty of Physics and Earth SciencesUniversity of Leipzig04103LeipzigGermany
- Institute of Clinical ImmunologyUniversity of Leipzig Medical School04103LeipzigGermany
- Dhirubhai Ambani Institute of Information and Communication TechnologyGandhinagar382 007India
| | - Adrian Keller
- Technical and Macromolecular ChemistryPaderborn UniversityWarburger Str. 10033098PaderbornGermany
| |
Collapse
|
41
|
Gbian DL, Omri A. Current and novel therapeutic strategies for the management of cystic fibrosis. Expert Opin Drug Deliv 2021; 18:535-552. [PMID: 33426936 DOI: 10.1080/17425247.2021.1874343] [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] [Indexed: 10/22/2022]
Abstract
Introduction: Cystic fibrosis (CF), is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene and affects thousands of people throughout the world. Lung disease is the leading cause of death in CF patients. Despite the advances in treatments, the management of CF mainly targets symptoms. Recent CFTR modulators however target common mutations in patients, alleviating symptoms of CF. Unfortunately, there is still no approved treatments for patients with rare mutations to date.Areas covered: This paper reviews current treatments of CF that mitigate symptoms and target genetic defects. The use of gene and drug delivery systems such as viral or non-viral vectors and nano-compounds to enhance CFTR expression and the activity of antimicrobials against chronic pulmonary infections respectively, will also be discussed.Expert opinion: Nano-compounds tackle biological barriers to drug delivery and revitalize antimicrobials, anti-inflammatory drugs and even genes delivery to CF patients. Gene therapy and gene editing are of particular interest because they have the potential to directly target genetic defects. Nanoparticles should be formulated to more specifically target epithelial cells, and biofilms. Finally, the development of more potent gene vectors to increase the duration of gene expression and reduce inflammation is a promising strategy to eventually cure CF.
Collapse
Affiliation(s)
- Douweh Leyla Gbian
- The Novel Drug and Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Abdelwahab Omri
- The Novel Drug and Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
42
|
Yadav K, Singh D, Singh MR, Pradhan M. Multifaceted targeting of cationic liposomes via co-delivery of anti-IL-17 siRNA and corticosteroid for topical treatment of psoriasis. Med Hypotheses 2020; 145:110322. [DOI: 10.1016/j.mehy.2020.110322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/19/2020] [Accepted: 09/26/2020] [Indexed: 01/05/2023]
|
43
|
Obuobi S, Škalko-Basnet N. Nucleic Acid Hybrids as Advanced Antibacterial Nanocarriers. Pharmaceutics 2020; 12:E643. [PMID: 32650506 PMCID: PMC7408145 DOI: 10.3390/pharmaceutics12070643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022] Open
Abstract
Conventional antibiotic therapy is often challenged by poor drug penetration/accumulation at infection sites and poses a significant burden to public health. Effective strategies to enhance the therapeutic efficacy of our existing arsenal include the use of nanoparticulate delivery platforms to improve drug targeting and minimize adverse effects. However, these nanocarriers are often challenged by poor loading efficiency, rapid release and inefficient targeting. Nucleic acid hybrid nanocarriers are nucleic acid nanosystems complexed or functionalized with organic or inorganic materials. Despite their immense potential in antimicrobial therapy, they are seldom utilized against pathogenic bacteria. With the emergence of antimicrobial resistance and the associated complex interplay of factors involved in antibiotic resistance, nucleic acid hybrids represent a unique opportunity to deliver antimicrobials against resistant pathogens and to target specific genes that control virulence or resistance. This review provides an unbiased overview on fabricating strategies for nucleic acid hybrids and addresses the challenges of pristine oligonucleotide nanocarriers. We report recent applications to enhance pathogen targeting, binding and control drug release. As multifunctional next-generational antimicrobials, the challenges and prospect of these nanocarriers are included.
Collapse
Affiliation(s)
- Sybil Obuobi
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Universitetsveien 57, 9037 Tromsø, Norway;
| | | |
Collapse
|