1
|
de Lacerda Coriolano D, de Souza JB, Cavalcanti IDL, Cavalcanti IMF. Antibacterial Activity of Polymyxins Encapsulated in Nanocarriers Against Gram-Negative Bacteria. Indian J Microbiol 2024. [DOI: 10.1007/s12088-024-01392-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/11/2024] [Indexed: 01/04/2025] Open
|
2
|
Stepanova M, Levit M, Egorova T, Nashchekina Y, Sall T, Demyanova E, Guryanov I, Korzhikova-Vlakh E. Poly(2-Deoxy-2-Methacrylamido-D-Glucose)-Based Complex Conjugates of Colistin, Deferoxamine and Vitamin B12: Synthesis and Biological Evaluation. Pharmaceutics 2024; 16:1080. [PMID: 39204425 PMCID: PMC11359296 DOI: 10.3390/pharmaceutics16081080] [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: 07/18/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Growing resistance to traditional antibiotics poses a global threat to public health. In this regard, modification of known antibiotics, but with limited applications due to side effects, is one of the extremely promising approaches at present. In this study, we proposed the synthesis of novel complex polymeric conjugates of the peptide antibiotic colistin (CT). A biocompatible and water-soluble synthetic glycopolymer, namely, poly(2-deoxy-2-methacrylamido-D-glucose) (PMAG), was used as a polymer carrier. In addition to monoconjugates containing CT linked to PMAG by hydrolyzable and stable bonds, a set of complex conjugates also containing the siderophore deferoxamine (DFOA) and vitamin B12 was developed. The structures of the conjugates were confirmed by 1H NMR and FTIR-spectroscopy, while the compositions of conjugates were determined by UV-Vis spectrophotometry and HPLC analysis. The buffer media with pH 7.4, corresponding to blood or ileum pH, and 5.2, corresponding to the intestinal pH after ingestion or pH in the focus of inflammation, were used to study the release of CT. The resulting conjugates were examined for cytotoxicity and antimicrobial activity. All conjugates showed less cytotoxicity than free colistin. A Caco-2 cell permeability assay was carried out for complex conjugates to simulate the drug absorption in the intestine. In contrast to free CT, which showed very low permeability through the Caco-2 monolayer, the complex polymeric conjugates of vitamin B12 and CT provided significant transport. The antimicrobial activity of the conjugates depended on the conjugate composition. It was found that conjugates containing CT linked to the polymer by a hydrolyzable bond were found to be more active than conjugates with a non-hydrolyzable bond between CT and PMAG. Conjugates containing DFOA complexed with Fe3+ were characterized by enhanced antimicrobial activity against Pseudomonas aeruginosa compared to other conjugates.
Collapse
Affiliation(s)
- Mariia Stepanova
- Institute of Macromolecular Compounds of Russian Academy of Sciences, 199004 St. Petersburg, Russia; (M.S.); (M.L.)
| | - Mariia Levit
- Institute of Macromolecular Compounds of Russian Academy of Sciences, 199004 St. Petersburg, Russia; (M.S.); (M.L.)
| | - Tatiana Egorova
- Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.E.); (E.D.)
| | - Yulia Nashchekina
- Institute of Cytology of Russian Academy of Sciences, 194064 St. Petersburg, Russia;
| | - Tatiana Sall
- Institute of Experimental Medicine, 197022 St. Petersburg, Russia;
| | - Elena Demyanova
- Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.E.); (E.D.)
| | - Ivan Guryanov
- Institute of Chemistry, St. Petersburg State University, 198504 St. Petersburg, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Macromolecular Compounds of Russian Academy of Sciences, 199004 St. Petersburg, Russia; (M.S.); (M.L.)
| |
Collapse
|
3
|
Paramshetti S, Angolkar M, Talath S, Osmani RAM, Spandana A, Al Fatease A, Hani U, Ramesh KVRNS, Singh E. Unravelling the in vivo dynamics of liposomes: Insights into biodistribution and cellular membrane interactions. Life Sci 2024; 346:122616. [PMID: 38599316 DOI: 10.1016/j.lfs.2024.122616] [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: 10/12/2023] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
Liposomes, as a colloidal drug delivery system dating back to the 1960s, remain a focal point of extensive research and stand as a highly efficient drug delivery method. The amalgamation of technological and biological advancements has propelled their evolution, elevating them to their current status. The key attributes of biodegradability and biocompatibility have been instrumental in driving substantial progress in liposome development. Demonstrating a remarkable ability to surmount barriers in drug absorption, enhance stability, and achieve targeted distribution within the body, liposomes have become pivotal in pharmaceutical research. In this comprehensive review, we delve into the intricate details of liposomal drug delivery systems, focusing specifically on their pharmacokinetics and cell membrane interactions via fusion, lipid exchange, endocytosis etc. Emphasizing the nuanced impact of various liposomal characteristics, we explore factors such as lipid composition, particle size, surface modifications, charge, dosage, and administration routes. By dissecting the multifaceted interactions between liposomes and biological barriers, including the reticuloendothelial system (RES), opsonization, enhanced permeability and retention (EPR) effect, ATP-binding cassette (ABC) phenomenon, and Complement Activation-Related Pseudoallergy (CARPA) effect, we provide a deeper understanding of liposomal behaviour in vivo. Furthermore, this review addresses the intricate challenges associated with translating liposomal technology into practical applications, offering insights into overcoming these hurdles. Additionally, we provide a comprehensive analysis of the clinical adoption and patent landscape of liposomes across diverse biomedical domains, shedding light on their potential implications for future research and therapeutic developments.
Collapse
Affiliation(s)
- Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - K V R N S Ramesh
- Department of Pharmaceutics, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Ekta Singh
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
4
|
Xie S, Li Y, Cao W, Peng J, Huang K, Meng J, Li X. Dual-Responsive Nanogels with Cascaded Gentamicin Release and Lysosomal Escape to Combat Intracellular Small Colony Variants for Peritonitis and Sepsis Therapies. Adv Healthc Mater 2024; 13:e2303671. [PMID: 38416744 DOI: 10.1002/adhm.202303671] [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: 10/24/2023] [Revised: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Intracellular bacteria are the major cause of serious infections including sepsis and peritonitis, but face great challenges in fighting against the stubborn intracellular small colony variants (SCVs). Herein, the authors have developed nanogels (NGs) to destroy both planktonic bacteria and SCVs and eliminate excessive inflammations for peritonitis and sepsis therapies. Free gentamicin (GEN) and hydroxyapatite nanoparticles (NPs) with GEN loading and mannose grafts (mHAG) are inoculated into ε-polylysine NGs to obtain NG@G1-mHAG2 through crosslinking with phenylboronic acid and tannic acid. The H2O2 consumption after reaction with phenylboronic esters and the elimination of free radicals by tannic acid alleviates the escalated inflammatory status to promote sepsis therapy. After mannose-mediated uptake into macrophages, the acid-triggered degradation of mHAG NPs generates Ca2+ to destabilize lysosomes and the efficient lysosomal escape leads to reversion of hypometabolic SCVs into normal phenotype and their sensitivity to GEN. In a peritonitis mouse model, NG@G1-mHAG2 treatment provides strong and persistent bactericidal effects against both extracellular bacteria and intracellular SCVs and extends survival of peritonitis mice without apparent hepatomegaly, splenomegaly, pulmonary edema, and inflammatory cell infiltration. Thus, this study demonstrates a concise and versatile strategy to eliminate SCVs and relieve inflammatory storms for peritonitis and sepsis therapies without infection recurrence.
Collapse
Affiliation(s)
- Shuang Xie
- School of Life Science and Engineering, Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yu Li
- School of Life Science and Engineering, Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Wenxiong Cao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jiawen Peng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Kun Huang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jie Meng
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xiaohong Li
- School of Life Science and Engineering, Key Laboratory of Advanced Technologies of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
5
|
Viera Herrera C, O'Connor PM, Ratrey P, Paul Ross R, Hill C, Hudson SP. Anionic liposome formulation for oral delivery of thuricin CD, a potential antimicrobial peptide therapeutic. Int J Pharm 2024; 654:123918. [PMID: 38401875 PMCID: PMC7615751 DOI: 10.1016/j.ijpharm.2024.123918] [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: 12/18/2023] [Revised: 01/30/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
Thuricin CD is a two-peptide antimicrobial produced by Bacillus thuringiensis. Unlike previous antibiotics, it has shown narrow spectrum activity against Clostridioides difficile, a bacterium capable of causing infectious disease in the colon. However, peptide antibiotics have stability, solubility, and permeability problems that can affect their performance in vivo. This work focuses on the bioactivity and bioavailability of thuricin CD with a view to developing a formulation for delivery of active thuricin CD peptides through the gastrointestinal tract (GIT) for local delivery in the colon. The results indicate that thuricin CD is active at low concentrations only when both peptides are present. While thuricin CD was degraded by proteases and was unstable and poorly soluble in gastric fluid, it showed increased solubility in intestinal fluid, probably due to micelle encapsulation. Based on this, thuricin CD was encapsulated in anionic liposomes, which showed increased activity compared to the free peptide, maintained activity after exposure to pepsin in gastric fluid and intestinal fluid, was stable in suspension for over 21 days at room temperature and for 60 days at 4 °C, and exhibited no toxicity to epithelial intestinal cells. These findings suggest that an anionic lipid-based nano formulation may be a promising approach for local oral delivery of thuricin CD.
Collapse
Affiliation(s)
- Camila Viera Herrera
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland
| | - Paula M O'Connor
- Food Biosciences, Teagasc, Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland Cork, Cork, Ireland
| | - Poonam Ratrey
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland
| | - R Paul Ross
- APC Microbiome Ireland Cork, Cork, Ireland; School of Microbiology, University College Cork, College Road, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland Cork, Cork, Ireland; School of Microbiology, University College Cork, College Road, Cork, Ireland
| | - Sarah P Hudson
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland.
| |
Collapse
|
6
|
Chen Y, Jiang Y, Xue T, Cheng J. Strategies for the eradication of intracellular bacterial pathogens. Biomater Sci 2024; 12:1115-1130. [PMID: 38284808 DOI: 10.1039/d3bm01498c] [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: 01/30/2024]
Abstract
Intracellular pathogens affect a significant portion of world population and cause millions of deaths each year. They can invade host cells and survive inside them and are extremely resistant to immune systems and antibiotics. Current treatments have limitations, and therefore, new effective therapies are needed to combat this ongoing health challenge. Active research efforts have been made to develop many new strategies to eradicate these intracellular pathogens. In this review, we focus on the intracellular bacterial pathogens and first introduce several representative intracellular bacteria and the diseases they cause. We then discuss the challenges in eradicating these bacteria and summarize the current therapeutics for intracellular bacteria. Finally, recent advances in intracellular bacteria eradication are highlighted.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Yunjiang Jiang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518071, China
| | - Tianrui Xue
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Biomaterials and Drug Delivery Laboratory, School of Engineering, Westlake University, Hangzhou 310024, China
| |
Collapse
|
7
|
Chen J, Hu S, Sun M, Shi J, Zhang H, Yu H, Yang Z. Recent advances and clinical translation of liposomal delivery systems in cancer therapy. Eur J Pharm Sci 2024; 193:106688. [PMID: 38171420 DOI: 10.1016/j.ejps.2023.106688] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/23/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
The limitations of conventional cancer treatment are driving the emergence and development of nanomedicines. Research in liposomal nanomedicine for cancer therapy is rapidly increasing, opening up new horizons for cancer treatment. Liposomal nanomedicine, which focuses on targeted drug delivery to improve the therapeutic effect of cancer while reducing damage to normal tissues and cells, has great potential in the field of cancer therapy. This review aims to clarify the advantages of liposomal delivery systems in cancer therapy. We describe the recent understanding of spatiotemporal fate of liposomes in the organism after different routes of drug administration. Meanwhile, various types of liposome-based drug delivery systems that exert their respective advantages in cancer therapy while reducing side effects were discussed. Moreover, the combination of liposomal agents with other therapies (such as photodynamic therapy and photothermal therapy) has demonstrated enhanced tumor-targeting efficiency and therapeutic efficacy. Finally, the opportunities and challenges faced by the field of liposome nanoformulations for entering the clinical treatment of cancer are highlighted.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life Sciences, Jilin University, Changchun, China
| | - Siyuan Hu
- School of Life Sciences, Jilin University, Changchun, China
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun, China
| | - Jianan Shi
- School of Life Sciences, Jilin University, Changchun, China
| | - Huan Zhang
- School of Life Sciences, Jilin University, Changchun, China
| | - Hongmei Yu
- China-Japan Union Hospital, Jilin University, Changchun, China.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China.
| |
Collapse
|
8
|
Ma X, Tang W, Yang R. Bioinspired nanomaterials for the treatment of bacterial infections. NANO RESEARCH 2024; 17:691-714. [DOI: 10.1007/s12274-023-6283-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 01/04/2025]
|
9
|
Razei A, Javanbakht M, Hajizade A, Heiat M, Zhao S, Aghamollaei H, Saadati M, Khafaei M, Asadi M, Cegolon L, Keihan AH. Nano and microparticle drug delivery systems for the treatment of Brucella infections. Biomed Pharmacother 2023; 169:115875. [PMID: 37979375 DOI: 10.1016/j.biopha.2023.115875] [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: 04/05/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023] Open
Abstract
Nano-based drug delivery systems are increasingly used for diagnosis, prevention and treatment of several diseases, thanks to several beneficial properties, including the ability to target specific cells or organs, allowing to reduce treatment costs and side effects frequently associated with chemotherapeutic medications, thereby improving treatment compliance of patients. In the field of communicable diseases, especially those caused by intracellular bacteria, the delivery of antibiotics targeting specific cells is of critical importance to maximize their treatment efficacy. Brucella melitensis, an intracellular obligate bacterium surviving and replicating inside macrophages is hard to be eradicated, mainly because of the low ability of antibiotics to enter these phagocityc cells . Although different antibiotics regimens including gentamicin, doxycycline and rifampicin are in fact used against the Brucellosis, no efficient treatment has been attained yet, due to the intracellular life of the respective pathogen. Nano-medicines responding to environmental stimuli allow to maximize drug delivery targeting macropages, thereby boosting treatment efficacy. Several drug delivery nano-technologies, including solid lipid nanoparticles, liposomes, chitosan, niosomes, and their combinations with chitosan sodium alginate can be employed in combination of antibiotics to successfully eradicate Brucellosis infection from patients.
Collapse
Affiliation(s)
- Ali Razei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center,Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajizade
- Biology Research Centre, Faculty of Basic Sciences, Imam Hossain University, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China
| | - Hossien Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mojtaba Saadati
- Biology Research Centre, Faculty of Basic Sciences, Imam Hossain University, Tehran, Iran
| | - Mostafa Khafaei
- Human Genetics Research Center, Baqiyatallah Medical Science University, Tehran, Iran
| | - Mosa Asadi
- Nephrology and Urology Research Center,Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Luca Cegolon
- University of Trieste, Department of Medical, Surgical & Health Sciences, Trieste, Italy; University Health Agency Giuliano-Isontina (ASUGI), Public Health Department, Trieste, Italy
| | - Amir Homayoun Keihan
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Abla KK, Mehanna MM. The battle of lipid-based nanocarriers against blood-brain barrier: a critical review. J Drug Target 2023; 31:832-857. [PMID: 37577919 DOI: 10.1080/1061186x.2023.2247583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
Central nervous system integrity is the state of brain functioning across sensory, cognitive, emotional-social behaviors, and motor domains, allowing a person to realise his full potential. Thus, brain disorders seriously affect patients' quality of life. Efficient drug delivery to treat brain disorders remains a crucial challenge due to numerous brain barriers, particularly the blood-brain barrier (BBB), which greatly impacts the ultimate drug therapeutic efficacy. Lately, nanocarrier technology has made huge progress in overcoming these barriers by improving drug solubility, ameliorating its retention, reducing its toxicity, and targeting the encapsulated agents to different brain tissues. The current review primarily offers an overview of the different components of BBB and the progress, strategies, and contemporary applications of the nanocarriers, specifically lipid-based nanocarriers (LBNs), in treating various brain disorders.
Collapse
Affiliation(s)
- Kawthar K Abla
- Pharmaceutical Nanotechnology Research Lab, Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Mohammed M Mehanna
- Faculty of Pharmacy, Industrial Pharmacy Department, Alexandria University, Alexandria, Egypt
| |
Collapse
|
11
|
Cesaro A, Lin S, Pardi N, de la Fuente-Nunez C. Advanced delivery systems for peptide antibiotics. Adv Drug Deliv Rev 2023; 196:114733. [PMID: 36804008 PMCID: PMC10771258 DOI: 10.1016/j.addr.2023.114733] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Antimicrobial peptides (AMPs) hold promise as alternatives to traditional antibiotics for preventing and treating multidrug-resistant infections. Although they have potent antimicrobial efficacy, AMPs are mainly limited by their susceptibility to proteases and potential off-site cytotoxicity. Designing the right delivery system for peptides can help to overcome such limitations, thus improving the pharmacokinetic and pharmacodynamic profiles of these drugs. The versatility of peptides and their genetically encodable structure make them suitable for both conventional and nucleoside-based formulations. In this review, we describe the main drug delivery procedures developed so far for peptide antibiotics: lipid nanoparticles, polymeric nanoparticles, hydrogels, functionalized surfaces, and DNA- and RNA-based delivery systems.
Collapse
Affiliation(s)
- Angela Cesaro
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Shuangzhe Lin
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
12
|
Karnwal A, Kumar G, Pant G, Hossain K, Ahmad A, Alshammari MB. Perspectives on Usage of Functional Nanomaterials in Antimicrobial Therapy for Antibiotic-Resistant Bacterial Infections. ACS OMEGA 2023; 8:13492-13508. [PMID: 37091369 PMCID: PMC10116640 DOI: 10.1021/acsomega.3c00110] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/28/2023] [Indexed: 05/03/2023]
Abstract
The clinical applications of nanotechnology are emerging as widely popular, particularly as a potential treatment approach for infectious diseases. Diseases associated with multiple drug-resistant organisms (MDROs) are a global concern of morbidity and mortality. The prevalence of infections caused by antibiotic-resistant bacterial strains has increased the urgency associated with researching and developing novel bactericidal medicines or unorthodox methods capable of combating antimicrobial resistance. Nanomaterial-based treatments are promising for treating severe bacterial infections because they bypass antibiotic resistance mechanisms. Nanomaterial-based approaches, especially those that do not rely on small-molecule antimicrobials, display potential since they can bypass drug-resistant bacteria systems. Nanoparticles (NPs) are small enough to pass through the cell membranes of pathogenic bacteria and interfere with essential molecular pathways. They can also target biofilms and eliminate infections that have proven difficult to treat. In this review, we described the antibacterial mechanisms of NPs against bacteria and the parameters involved in targeting established antibiotic resistance and biofilms. Finally, yet importantly, we talked about NPs and the various ways they can be utilized, including as delivery methods, intrinsic antimicrobials, or a mixture.
Collapse
Affiliation(s)
- Arun Karnwal
- Department
of Microbiology, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Gaurav Kumar
- Department
of Microbiology, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Gaurav Pant
- Department
of Microbiology, Graphic Era (Deemed to
be University), Dehradun, Uttarakhand 248002, India
| | - Kaizar Hossain
- Department
of Environmental Science, Asutosh College, University of Calcutta, 92, Shyama Prasad Mukherjee Road, Bhowanipore, Kolkata 700026, West
Bengal, India
| | - Akil Ahmad
- Department
of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammed B. Alshammari
- Department
of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
13
|
Karpuz M, Temel A, Ozgenc E, Tekintas Y, Erel-Akbaba G, Senyigit Z, Atlihan-Gundogdu E. 99mTc-Labeled, Colistin Encapsulated, Theranostic Liposomes for Pseudomonas aeruginosa Infection. AAPS PharmSciTech 2023; 24:77. [PMID: 36899198 DOI: 10.1208/s12249-023-02533-8] [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: 10/20/2022] [Accepted: 02/12/2023] [Indexed: 03/12/2023] Open
Abstract
Infectious diseases are still the major issue not only due to antibiotic resistance but also causing deaths if not diagnosed at early-stages. Different approaches including nanosized drug delivery systems and theranostics are researched to overcome antibiotic resistance, decrease the side effects of antibiotics, improve the treatment response, and early diagnose. Therefore, in the present study, nanosized, radiolabeled with 99mTc, colistin encapsulated, neutral and cationic liposome formulations were prepared as the theranostic agent for Pseudomonas aeruginosa infections. Liposomes exhibited appropriate physicochemical properties thanks to their nano-particle size (between 173 and 217 nm), neutral zeta potential value (about - 6.5 and 2.8 mV), as well as encapsulation efficiency of about 75%. All liposome formulations were radiolabeled with over 90% efficiency, and the concentration of stannous chloride was found as 1 mg.mL-1 to obtain maximum radiolabeling efficiency. In alamar blue analysis, neutral liposome formulations were found more biocompatible compared with the cationic formulations. Neutral colistin encapsulated liposomes were found to be more effective against P. aeruginosa strain according to their time-dependent antibacterial effect, in addition to their highest bacterial binding capacity. As conclusion, theranostic, nanosized, colistin encapsulated, neutral liposome formulations were found as promising agents for the imaging and treating of P. aeruginosa infections.
Collapse
Affiliation(s)
- Merve Karpuz
- Department of Radiopharmacy, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey.
| | - Aybala Temel
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Emre Ozgenc
- Department of Radiopharmacy, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Yamac Tekintas
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Gulsah Erel-Akbaba
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Zeynep Senyigit
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | | |
Collapse
|
14
|
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
|
15
|
Hetta HF, Ramadan YN, Al-Harbi AI, A. Ahmed E, Battah B, Abd Ellah NH, Zanetti S, Donadu MG. Nanotechnology as a Promising Approach to Combat Multidrug Resistant Bacteria: A Comprehensive Review and Future Perspectives. Biomedicines 2023; 11:biomedicines11020413. [PMID: 36830949 PMCID: PMC9953167 DOI: 10.3390/biomedicines11020413] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
The wide spread of antibiotic resistance has been alarming in recent years and poses a serious global hazard to public health as it leads to millions of deaths all over the world. The wide spread of resistance and sharing resistance genes between different types of bacteria led to emergence of multidrug resistant (MDR) microorganisms. This problem is exacerbated when microorganisms create biofilms, which can boost bacterial resistance by up to 1000-fold and increase the emergence of MDR infections. The absence of novel and potent antimicrobial compounds is linked to the rise of multidrug resistance. This has sparked international efforts to develop new and improved antimicrobial agents as well as innovative and efficient techniques for antibiotic administration and targeting. There is an evolution in nanotechnology in recent years in treatment and prevention of the biofilm formation and MDR infection. The development of nanomaterial-based therapeutics, which could overcome current pathways linked to acquired drug resistance, is a hopeful strategy for treating difficult-to-treat bacterial infections. Additionally, nanoparticles' distinct size and physical characteristics enable them to target biofilms and treat resistant pathogens. This review highlights the current advances in nanotechnology to combat MDR and biofilm infection. In addition, it provides insight on development and mechanisms of antibiotic resistance, spread of MDR and XDR infection, and development of nanoparticles and mechanisms of their antibacterial activity. Moreover, this review considers the difference between free antibiotics and nanoantibiotics, and the synergistic effect of nanoantibiotics to combat planktonic bacteria, intracellular bacteria and biofilm. Finally, we will discuss the strength and limitations of the application of nanotechnology against bacterial infection and future perspectives.
Collapse
Affiliation(s)
- Helal F. Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
- Correspondence: (H.F.H.); (M.G.D.)
| | - Yasmin N. Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Alhanouf I. Al-Harbi
- Department of Medical Laboratory, College of Applied Medical Sciences, Taibah University, Yanbu 46411, Saudi Arabia
| | - Esraa A. Ahmed
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Basem Battah
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Syrian Private University (SPU), Daraa International Highway, 36822 Damascus, Syria
| | - Noura H. Abd Ellah
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, Badr University in Assiut, Naser City, Assiut 2014101, Egypt
| | - Stefania Zanetti
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Matthew Gavino Donadu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Hospital Pharmacy, Azienda Ospedaliero Universitaria di Sassari, 07100 Sassari, Italy
- Correspondence: (H.F.H.); (M.G.D.)
| |
Collapse
|
16
|
van Gent ME, van Baaren T, Kłodzińska SN, Ali M, Dolezal N, van Doodewaerd BR, Bos E, de Waal AM, Koning RI, Drijfhout JW, Nielsen HM, Nibbering PH. Encapsulation of SAAP-148 in Octenyl Succinic Anhydride-Modified Hyaluronic Acid Nanogels for Treatment of Skin Wound Infections. Pharmaceutics 2023; 15:pharmaceutics15020429. [PMID: 36839751 PMCID: PMC9967827 DOI: 10.3390/pharmaceutics15020429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Chronic wound infections colonized by bacteria are becoming more difficult to treat with current antibiotics due to the development of antimicrobial resistance (AMR) as well as biofilm and persister cell formation. Synthetic antibacterial and antibiofilm peptide (SAAP)-148 is an excellent alternative for treatment of such infections but suffers from limitations related to its cationic peptidic nature and thus instability and possible cytotoxicity, resulting in a narrow therapeutic window. Here, we evaluated SAAP-148 encapsulation in nanogels composed of octenyl succinic anhydride (OSA)-modified hyaluronic acid (HA) to circumvent these limitations. SAAP-148 was efficiently (>98%) encapsulated with high drug loading (23%), resulting in monodispersed anionic OSA-HA nanogels with sizes ranging 204-253 nm. Nanogel lyophilization in presence of polyvinyl alcohol maintained their sizes and morphology. SAAP-148 was sustainedly released from lyophilized nanogels (37-41% in 72 h) upon reconstitution. Lyophilized SAAP-148-loaded nanogels showed similar antimicrobial activity as SAAP-148 against planktonic and biofilm-residing AMR Staphylococcus aureus and Acinetobacter baumannii. Importantly, formulated SAAP-148 showed reduced cytotoxicity against human erythrocytes, primary human skin fibroblasts and human keratinocytes. Additionally, lyophilized SAAP-148-loaded nanogels eradicated AMR S. aureus and A. baumannii colonizing a 3D human epidermal model, without inducing any cytotoxicity in contrast to SAAP-148. These findings indicate that OSA-HA nanogels increase SAAP-148's therapeutic potential for treatment of skin wound infections.
Collapse
Affiliation(s)
- Miriam E. van Gent
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence:
| | - Tom van Baaren
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sylvia N. Kłodzińska
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Muhanad Ali
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Natasja Dolezal
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Bjorn R. van Doodewaerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Erik Bos
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Amy M. de Waal
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Roman I. Koning
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Hanne Mørck Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
17
|
Wang C, Yang Y, Cao Y, Liu K, Shi H, Guo X, Liu W, Hao R, Song H, Zhao R. Nanocarriers for the delivery of antibiotics into cells against intracellular bacterial infection. Biomater Sci 2023; 11:432-444. [PMID: 36503914 DOI: 10.1039/d2bm01489k] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The barrier function of host cells enables intracellular bacteria to evade the lethality of the host immune system and antibiotics, thereby causing chronic and recurrent infections that seriously threaten human health. Currently, the main clinical strategy for the treatment of intracellular bacterial infections involves the use of long-term and high-dose antibiotics. However, insufficient intracellular delivery of antibiotics along with various resistance mechanisms not only weakens the efficacy of current therapies but also causes serious adverse drug reactions, further increasing the disease and economic burden. Improving the delivery efficiency, intracellular accumulation, and action time of antibiotics remains the most economical and effective way to treat intracellular bacterial infections. The rapid development of nanotechnology provides a strategy to efficiently deliver antibiotics against intracellular bacterial infections into cells. In this review, we summarize the types of common intracellular pathogens, the difficulties faced by antibiotics in the treatment of intracellular bacterial infections, and the research progress of several types of representative nanocarriers for the delivery of antibiotics against intracellular bacterial infections that have emerged in recent years. This review is expected to provide a reference for further elucidating the intracellular transport mechanism of nanocarrier-drug complexes, designing safer and more effective nanocarriers and establishing new strategies against intracellular bacterial infection.
Collapse
Affiliation(s)
- Chao Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yi Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Kaixin Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Hua Shi
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Xudong Guo
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Wanying Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Hongbin Song
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongtao Zhao
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| |
Collapse
|
18
|
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: 6] [Impact Index Per Article: 3.0] [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
|
19
|
Brar A, Majumder S, Navarro MZ, Benoit-Biancamano MO, Ronholm J, George S. Nanoparticle-Enabled Combination Therapy Showed Superior Activity against Multi-Drug Resistant Bacterial Pathogens in Comparison to Free Drugs. NANOMATERIALS 2022; 12:nano12132179. [PMID: 35808015 PMCID: PMC9268018 DOI: 10.3390/nano12132179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023]
Abstract
The emergence of multidrug-resistant (MDR) bacterial pathogens in farm animals and their zoonotic spread is a concern to both animal agriculture and public health. Apart from antimicrobial resistance (AMR), bacterial pathogens from the genera of Salmonella and Staphylococcus take refuge inside host cells, thereby demanding intervention strategies that can eliminate intracellular MDR pathogens. In this study, seven clinical isolates of Salmonella and Staphylococcus from swine farms were characterized for antibiotic (n = 24) resistance, resistance mechanisms, and virulence characteristics. All isolates showed resistance to one or more antibiotics and S. enterica ser. Typhimurium isolate had the highest resistance to the panel of antibiotics tested. Major resistance mechanisms identified were efflux pump and beta-lactamase enzyme activities. Staphylococcus isolates showed complete hemolysis and strong biofilm formation, while Salmonella isolates caused partial hemolysis, but showed no or weak biofilm formation. MDR isolates of S. aureus M12 and S. enterica ser. Typhimurium bacteria were subsequently tested against combinations of antibiotics and potentiating adjuvants for improved antibacterial efficacy using a checkerboard assay, and their fractional inhibitory concentration index (FICI) was calculated. A combination of chitosan and silica nanoparticles containing tetracycline (TET) and efflux pump inhibitor chlorpromazine (CPZ), respectively, was characterized for physicochemical properties and effectiveness against MDR Salmonella enterica ser. Typhimurium isolate. This combination of nano-encapsulated drugs improved the antibacterial efficacy by inhibiting AMR mechanisms (efflux activity, beta-lactamase enzyme activity, and hydrogen sulfide (H2S) production) and reducing intracellular pathogen load by 83.02 ± 14.35%. In conclusion, this study sheds light on the promising applicability of nanoparticle-enabled combination therapy to combat multidrug-resistant pathogens encountered in animal agriculture.
Collapse
Affiliation(s)
- Amarpreet Brar
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, 21111 Lakeshore, Ste Anne de Bellevue, QC H9X 3V9, Canada; (A.B.); (S.M.); (J.R.)
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, QC J2S 2M2, Canada; (M.Z.N.); (M.-O.B.-B.)
| | - Satwik Majumder
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, 21111 Lakeshore, Ste Anne de Bellevue, QC H9X 3V9, Canada; (A.B.); (S.M.); (J.R.)
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, QC J2S 2M2, Canada; (M.Z.N.); (M.-O.B.-B.)
| | - Maria Zardon Navarro
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, QC J2S 2M2, Canada; (M.Z.N.); (M.-O.B.-B.)
- Research Group on Infectious Diseases in Production Animals (GREMIP), Department of Pathology and Microbiology, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Marie-Odile Benoit-Biancamano
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, QC J2S 2M2, Canada; (M.Z.N.); (M.-O.B.-B.)
- Research Group on Infectious Diseases in Production Animals (GREMIP), Department of Pathology and Microbiology, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Jennifer Ronholm
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, 21111 Lakeshore, Ste Anne de Bellevue, QC H9X 3V9, Canada; (A.B.); (S.M.); (J.R.)
- Department of Animal Science, Macdonald Campus, McGill University, 2111 Lakeshore, Ste Anne de Bellevue, QC H9X 3V9, Canada
| | - Saji George
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, 21111 Lakeshore, Ste Anne de Bellevue, QC H9X 3V9, Canada; (A.B.); (S.M.); (J.R.)
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, QC J2S 2M2, Canada; (M.Z.N.); (M.-O.B.-B.)
- Correspondence: ; Tel.: +1-514-398-7920; Fax: +1-514-398-7990
| |
Collapse
|
20
|
Huck BC, Thiyagarajan D, Bali A, Boese A, Besecke KFW, Hozsa C, Gieseler RK, Furch M, Carvalho‐Wodarz C, Waldow F, Schwudke D, Metelkina O, Titz A, Huwer H, Schwarzkopf K, Hoppstädter J, Kiemer AK, Koch M, Loretz B, Lehr C. Nano-in-Microparticles for Aerosol Delivery of Antibiotic-Loaded, Fucose-Derivatized, and Macrophage-Targeted Liposomes to Combat Mycobacterial Infections: In Vitro Deposition, Pulmonary Barrier Interactions, and Targeted Delivery. Adv Healthc Mater 2022; 11:e2102117. [PMID: 35112802 PMCID: PMC11468583 DOI: 10.1002/adhm.202102117] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/14/2022] [Indexed: 12/12/2022]
Abstract
Nontuberculous mycobacterial infections rapidly emerge and demand potent medications to cope with resistance. In this context, targeted loco-regional delivery of aerosol medicines to the lungs is an advantage. However, sufficient antibiotic delivery requires engineered aerosols for optimized deposition. Here, the effect of bedaquiline-encapsulating fucosylated versus nonfucosylated liposomes on cellular uptake and delivery is investigated. Notably, this comparison includes critical parameters for pulmonary delivery, i.e., aerosol deposition and the noncellular barriers of pulmonary surfactant (PS) and mucus. Targeting increases liposomal uptake into THP-1 cells as well as peripheral blood monocyte- and lung-tissue derived macrophages. Aerosol deposition in the presence of PS, however, masks the effect of active targeting. PS alters antibiotic release that depends on the drug's hydrophobicity, while mucus reduces the mobility of nontargeted more than fucosylated liposomes. Dry-powder microparticles of spray-dried bedaquiline-loaded liposomes display a high fine particle fraction of >70%, as well as preserved liposomal integrity and targeting function. The antibiotic effect is maintained when deposited as powder aerosol on cultured Mycobacterium abscessus. When treating M. abscessus infected THP-1 cells, the fucosylated variant enabled enhanced bacterial killing, thus opening up a clear perspective for the improved treatment of nontuberculous mycobacterial infections.
Collapse
Affiliation(s)
- Benedikt C. Huck
- Department of Drug DeliveryHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Department of PharmacyHelmholtz Institute for Pharmaceutical Research SaarlandSaarland UniversityCampus E8 1Saarbrücken66123Germany
| | - Durairaj Thiyagarajan
- Department of Anti‐infective Drug DiscoveryHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8 1Saarbrücken66123Germany
| | - Aghiad Bali
- Department of Drug DeliveryHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Department of PharmacyHelmholtz Institute for Pharmaceutical Research SaarlandSaarland UniversityCampus E8 1Saarbrücken66123Germany
| | - Annette Boese
- Department of Drug DeliveryHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Karen F. W. Besecke
- Rodos Biotarget GmbHHannover30625Germany
- Present address:
Solmic BioTech GmbHDüsseldorf40225Germany
| | - Constantin Hozsa
- Rodos Biotarget GmbHHannover30625Germany
- Present address:
Siegfried AG HamelnHameln31789Germany
| | - Robert K. Gieseler
- Rodos Biotarget GmbHHannover30625Germany
- Laboratory of Immunology and Molecular Biologyand Department of Internal MedicineUniversity HospitalKnappschaftskrankenhaus BochumRuhr University BochumBochum44892Germany
| | - Marcus Furch
- Rodos Biotarget GmbHHannover30625Germany
- Present address:
Biolife Holding AGHeidelberg69126Germany
| | - Cristiane Carvalho‐Wodarz
- Department of Drug DeliveryHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Franziska Waldow
- Research Center BorstelLeibniz Lung CenterBorstel23845Germany
- German Center for Infection ResearchThematic Translational Unit TuberculosisPartner Site Hamburg‐Lübeck‐Borstel‐RiemsBraunschweig38124Germany
| | - Dominik Schwudke
- Research Center BorstelLeibniz Lung CenterBorstel23845Germany
- German Center for Infection ResearchThematic Translational Unit TuberculosisPartner Site Hamburg‐Lübeck‐Borstel‐RiemsBraunschweig38124Germany
- German Center for Lung Research (DZL)Airway Research Center North (ARCN)Kiel NanoSurface and Interface Science KiNSISKiel UniversityKiel24118Germany
| | - Olga Metelkina
- Chemical Biology of Carbohydrates (CBCH)Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Center for Infection ResearchSaarbrücken66123Germany
- Department of ChemistrySaarland UniversitySaarbrücken66123Germany
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH)Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Center for Infection ResearchSaarbrücken66123Germany
- Department of ChemistrySaarland UniversitySaarbrücken66123Germany
- Deutsches Zentrum für Infektionsforschung (DZIF)Hannover‐Braunschweig siteBraunschweig38124Germany
| | - Hanno Huwer
- Cardiothoracic SurgeryHeart Center VoelklingenVölklingen66333Germany
| | - Konrad Schwarzkopf
- Department of Anaesthesia and Intensive CareKlinikum Saarbrücken gGmbHSaarbrücken66119Germany
| | | | | | - Marcus Koch
- INM – Leibniz Institute for New MaterialsCampus D2 2Saarbrücken66123Germany
| | - Brigitta Loretz
- Department of Drug DeliveryHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
| | - Claus‐Michael Lehr
- Department of Drug DeliveryHelmholtz Institute for Pharmaceutical Research SaarlandCampus E8.1Saarbrücken66123Germany
- Department of PharmacyHelmholtz Institute for Pharmaceutical Research SaarlandSaarland UniversityCampus E8 1Saarbrücken66123Germany
| |
Collapse
|
21
|
Sheikholeslami B, Lam NW, Dua K, Haghi M. Exploring the impact of physicochemical properties of liposomal formulations on their in vivo fate. Life Sci 2022; 300:120574. [DOI: 10.1016/j.lfs.2022.120574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 12/16/2022]
|
22
|
Deng K, Li Y, Liang X, Shen C, Zeng Z, Xu X. Virus-inspired nanoparticles as versatile antibacterial carriers for antibiotic delivery against Gram-negative and Gram-positive bacteria. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.09.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
23
|
Current Advances in Lipid and Polymeric Antimicrobial Peptide Delivery Systems and Coatings for the Prevention and Treatment of Bacterial Infections. Pharmaceutics 2021; 13:pharmaceutics13111840. [PMID: 34834254 PMCID: PMC8618997 DOI: 10.3390/pharmaceutics13111840] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Bacterial infections constitute a threat to public health as antibiotics are becoming less effective due to the emergence of antimicrobial resistant strains and biofilm and persister formation. Antimicrobial peptides (AMPs) are considered excellent alternatives to antibiotics; however, they suffer from limitations related to their peptidic nature and possible toxicity. The present review critically evaluates the chemical characteristics and antibacterial effects of lipid and polymeric AMP delivery systems and coatings that offer the promise of enhancing the efficacy of AMPs, reducing their limitations and prolonging their half-life. Unfortunately, the antibacterial activities of these systems and coatings have mainly been evaluated in vitro against planktonic bacteria in less biologically relevant conditions, with only some studies focusing on the antibiofilm activities of the formulated AMPs and on the antibacterial effects in animal models. Further improvements of lipid and polymeric AMP delivery systems and coatings may involve the functionalization of these systems to better target the infections and an analysis of the antibacterial activities in biologically relevant environments. Based on the available data we proposed which polymeric AMP delivery system or coatings could be profitable for the treatment of the different hard-to-treat infections, such as bloodstream infections and catheter- or implant-related infections.
Collapse
|
24
|
Subramaniam S, Joyce P, Thomas N, Prestidge CA. Bioinspired drug delivery strategies for repurposing conventional antibiotics against intracellular infections. Adv Drug Deliv Rev 2021; 177:113948. [PMID: 34464665 DOI: 10.1016/j.addr.2021.113948] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/04/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022]
Abstract
Bacteria have developed a wealth of strategies to avoid and resist the action of antibiotics, one of which involves pathogens invading and forming reservoirs within host cells. Due to the poor cell membrane permeability, stability and retention of conventional antibiotics, this renders current treatments largely ineffective, since achieving a therapeutically relevant antibiotic concentration at the site of intracellular infection is not possible. To overcome such challenges, current antibiotics are 'repurposed' via reformulation using micro- or nano-carrier systems that effectively encapsulate and deliver therapeutics across cellular membranes of infected cells. Bioinspired materials that imitate the uptake of biological particulates and release antibiotics in response to natural stimuli are recently explored to improve the targeting and specificity of this 'nanoantibiotic' approach. In this review, the mechanisms of internalization and survival of intracellular bacteria are elucidated, effectively accentuating the current treatment challenges for intracellular infections and the implications for repurposing conventional antibiotics. Key case studies of nanoantibiotics that have drawn inspiration from natural biological particles and cellular uptake pathways to effectively eradicate intracellular pathogens are detailed, clearly highlighting the rational for harnessing bioinspired drug delivery strategies.
Collapse
Affiliation(s)
- Santhni Subramaniam
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia
| | - Paul Joyce
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia
| | - Nicky Thomas
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia; The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia
| | - Clive A Prestidge
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
25
|
Lin L, Chi J, Yan Y, Luo R, Feng X, Zheng Y, Xian D, Li X, Quan G, Liu D, Wu C, Lu C, Pan X. Membrane-disruptive peptides/peptidomimetics-based therapeutics: Promising systems to combat bacteria and cancer in the drug-resistant era. Acta Pharm Sin B 2021; 11:2609-2644. [PMID: 34589385 PMCID: PMC8463292 DOI: 10.1016/j.apsb.2021.07.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 02/05/2023] Open
Abstract
Membrane-disruptive peptides/peptidomimetics (MDPs) are antimicrobials or anticarcinogens that present a general killing mechanism through the physical disruption of cell membranes, in contrast to conventional chemotherapeutic drugs, which act on precise targets such as DNA or specific enzymes. Owing to their rapid action, broad-spectrum activity, and mechanisms of action that potentially hinder the development of resistance, MDPs have been increasingly considered as future therapeutics in the drug-resistant era. Recently, growing experimental evidence has demonstrated that MDPs can also be utilized as adjuvants to enhance the therapeutic effects of other agents. In this review, we evaluate the literature around the broad-spectrum antimicrobial properties and anticancer activity of MDPs, and summarize the current development and mechanisms of MDPs alone or in combination with other agents. Notably, this review highlights recent advances in the design of various MDP-based drug delivery systems that can improve the therapeutic effect of MDPs, minimize side effects, and promote the co-delivery of multiple chemotherapeutics, for more efficient antimicrobial and anticancer therapy.
Collapse
Affiliation(s)
- Liming Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Jiaying Chi
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Yilang Yan
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Rui Luo
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xiaoqian Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Yuwei Zheng
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Dongyi Xian
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xin Li
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Daojun Liu
- Shantou University Medical College, Shantou 515041, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Chao Lu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
26
|
Shakouri A, Kahroba H, Hamishekar H, Abdolalizadeh J. Nanoencapsulation of Hirudo medicinalis proteins in liposomes as a nanocarrier for inhibiting angiogenesis through targeting VEGFA in the Breast cancer cell line (MCF-7). BIOIMPACTS 2021; 12:115-126. [PMID: 35411300 PMCID: PMC8905592 DOI: 10.34172/bi.2021.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 11/09/2022]
Abstract
Introduction: Breast cancer is the most serious cause of women’s death throughout the world. Using nanocarrier vehicles to the exact site of cancer upgrades the therapeutic efficiency of the drugs. Capsulation of active proteins in the vesicular liposomes’ hydrophilic core is essential to develop a therapeutic protein carrier system. We aimed to encapsulate the medicinal leech saliva extract (LSE) and assess the inhibition of angiogenesis of breast cancer cells by targeting vascular endothelial growth factor A (VEGFA). Methods: In this research, enhanced formulation of liposomal protein was determined by zeta potential analysis, droplet size, drug release assay, and transmission electron microscopy (TEM). Furthermore, a cytotoxicity assay of liposomal LSE was performed to determine the cytotoxic activity of components. For assessing the expression of VEGFA, P53, and hypoxia-inducible factor subunit alpha (HIF1a) genes, Real-Time PCR was applied. Results: Nano liposome was chosen as an enhanced formulation due to its much smaller size (46.23 nm). Liposomal LSE had more practical actions on the MCF-7 cells. As noticed by DAPI staining, apoptosis was extensively greater in treated MCF-7 cells. Wound healing assay demonstrated that MCF-7 cells could not sustain growth at the presence of liposomal LSE and expression of the VEGFA gene was declined in treated cells. Downregulation of VEGFA was evaluated with western blotting technique. Conclusion: It can be concluded that our investigation of the tests confirmed the fact that nano liposomal LSE is a novel promising formulation for anticancer drugs and can significantly improve the penetration of protein drugs to cancer cells.
Collapse
Affiliation(s)
- Amir Shakouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Houman Kahroba
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishekar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
He Y, Ayansola H, Hou Q, Liao C, Lei J, Lai Y, Jiang Q, Masatoshi H, Zhang B. Genistein Inhibits Colonic Goblet Cell Loss and Colorectal Inflammation Induced by Salmonella Typhimurium Infection. Mol Nutr Food Res 2021; 65:e2100209. [PMID: 34146390 DOI: 10.1002/mnfr.202100209] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/12/2021] [Indexed: 12/28/2022]
Abstract
SCOPE Salmonella is the main food-borne pathogen, which can infect intestinal epithelial cells and causes colitis. Genistein has a variety of biological activities that alleviates colitis induced by sodium dextran sulfate in a variety of ways, but its protective effects on colitis caused by pathogenic bacteria are still unknown. METHODS AND RESULTS This study explores the protective effect of genistein in reducing colitis caused by Salmonella infection. Salmonella causes colon inflammation through activating cyclooxygenase-2/prostaglandin E2, and genistein inhibits colitis caused by Salmonella typhimurium infection. Salmonella infection increases colonic mucosal damage, proliferating cells, and goblet cell loss, while the administration of genistein solves these pathological changes. In addition, it is further proved that Salmonella causes severe colitis related to goblet cell loss and activates the host crypt stem cells to repair the damaged epithelium. Salmonella infection inhibites the host mammalian target of rapamycin, activates light chain 3 II pathways to induce autophagy to eliminate pathogenic bacteria. Genistein increases Lactobacillus in feces and reduces Salmonella colonization to inhibit colitis induces by Salmonella infection. CONCLUSION This study demonstrates genistein alleviated colitis and inhibites the goblet cell loss causes by Salmonella infection through regulating the gut bacteria and intestinal stem cell development.
Collapse
Affiliation(s)
- Yang He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
- College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Hammed Ayansola
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Qihang Hou
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Chaoyong Liao
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Jiaqi Lei
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Yujiao Lai
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Qiuyu Jiang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Hori Masatoshi
- Department of Veterinary Pharmacology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| |
Collapse
|
28
|
Antezana PE, Municoy S, Bellino MG, Martini MF, Desimone MF. Nanodelivery of the Gramicidin Peptide for Enhancing Antimicrobial Activity. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Pablo E. Antezana
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química y Metabolismo del Fármaco (IQUIMEFA) Facultad de Farmacia y Bioquímica Junín 956, (1113) Buenos Aires Argentina
| | - Sofia Municoy
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química y Metabolismo del Fármaco (IQUIMEFA) Facultad de Farmacia y Bioquímica Junín 956, (1113) Buenos Aires Argentina
| | - Martín G. Bellino
- Instituto de Nanociencia y Nanotecnología – Comisión Nacional de Energía Atómica y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Av. General Paz 1499, (1650) San Martín Argentina
| | - M. Florencia Martini
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química y Metabolismo del Fármaco (IQUIMEFA) Facultad de Farmacia y Bioquímica Junín 956, (1113) Buenos Aires Argentina
| | - Martín F. Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química y Metabolismo del Fármaco (IQUIMEFA) Facultad de Farmacia y Bioquímica Junín 956, (1113) Buenos Aires Argentina
| |
Collapse
|
29
|
Jash A, Ubeyitogullari A, Rizvi SSH. Liposomes for oral delivery of protein and peptide-based therapeutics: challenges, formulation strategies, and advances. J Mater Chem B 2021; 9:4773-4792. [PMID: 34027542 DOI: 10.1039/d1tb00126d] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Throughout the past decade, there has been a rapid growth in the development of protein/peptide-based therapeutics. These therapeutics have found widespread applications in the treatment of cancer, infectious diseases, and other metabolic disorders owing to their several desirable attributes, such as reduced toxicity, diverse biological activities, high specificity, and potency. Most protein/peptide-based drugs are still administered parenterally, and there is an unprecedented demand in the pharmaceutical industry to develop oral delivery routes to increase patient acceptability and convenience. Recent advancements in nanomedicine discoveries have led to the development of several nano and micro-particle-based oral delivery platforms for protein/peptide-based therapeutics and among these, liposomes have emerged as a prominent candidate. Liposomes are spherical vesicles composed of one or more phospholipid bilayers enclosing a core aqueous phase. Their unique amphiphilic nature enables encapsulation of a diverse range of bioactives/drugs including both hydrophobic and hydrophilic compounds for delivery. Against this backdrop, this review provides an overview of the current approaches and challenges associated with the routes and methods of oral administration of protein/peptide-based therapeutics by using liposomes as a potential vehicle. First, the conventional and innovative liposome formation approaches have been discussed along with their applications. Next, the challenges associated with current approaches for oral delivery of protein and peptide-derived therapeutics have been thoroughly addressed. Lastly, we have critically reviewed the potential of liposomes utilization as vehicles for oral delivery of proteins emphasizing the current status and future directions in this area.
Collapse
Affiliation(s)
- Apratim Jash
- Department of Food Science, Cornell University, Ithaca, NY 14850, USA
| | | | | |
Collapse
|
30
|
Khan MA. Targeted Drug Delivery Using Tuftsin-bearing Liposomes: Implications in the Treatment of Infectious Diseases and Tumors. Curr Drug Targets 2021; 22:770-778. [PMID: 33243117 DOI: 10.2174/1389450121999201125200756] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/04/2020] [Accepted: 10/13/2020] [Indexed: 11/22/2022]
Abstract
Tuftsin, a tetrapeptide (Thr-Lys-Pro-Arg), acts as an immunopotentiating molecule with its ability to bind and activate many immune cells, including macrophages or monocytes, neutrophils and dendritic cells. The specific targeting activity of tuftsin has been further increased by its palmitoylation followed by its incorporation into the lipid bilayer of liposomes. Tuftsin-bearing liposomes (Tuft-liposomes) possess several characteristics that enable them to act as a potential drug and vaccine carriers. Tuft-liposomes-loaded anti-microbial drugs have been shown to be highly effective against many infectious diseases, including tuberculosis, leishmaniasis, malaria, candidiasis and cryptococosis. Moreover, Tuft-liposomes also increased the activity of anticancer drug etoposide against fibrosarcoma in mice. Tuft-liposomes showed the immune-potentiating effect and rejuvenated the immune cells in the leukopenic mice. In addition, antigens encapsulated in Tuftsin-bearing liposomes demonstrated greater immunogenicity by increasing the T cell proliferation and antibody secretion. Keeping into consideration their specific targeting and immunopotentiating effects, Tuft-liposomes may potentially be used as promising drug and vaccine delivery systems.
Collapse
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
31
|
Arshad R, Pal K, Sabir F, Rahdar A, Bilal M, Shahnaz G, Kyzas GZ. A review of the nanomaterials use for the diagnosis and therapy of salmonella typhi. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.129928] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Makabenta JMV, Nabawy A, Li CH, Schmidt-Malan S, Patel R, Rotello VM. Nanomaterial-based therapeutics for antibiotic-resistant bacterial infections. Nat Rev Microbiol 2021; 19:23-36. [PMID: 32814862 PMCID: PMC8559572 DOI: 10.1038/s41579-020-0420-1] [Citation(s) in RCA: 563] [Impact Index Per Article: 140.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2020] [Indexed: 12/22/2022]
Abstract
Antibiotic-resistant bacterial infections arising from acquired resistance and/or through biofilm formation necessitate the development of innovative 'outside of the box' therapeutics. Nanomaterial-based therapies are promising tools to combat bacterial infections that are difficult to treat, featuring the capacity to evade existing mechanisms associated with acquired drug resistance. In addition, the unique size and physical properties of nanomaterials give them the capability to target biofilms, overcoming recalcitrant infections. In this Review, we highlight the general mechanisms by which nanomaterials can be used to target bacterial infections associated with acquired antibiotic resistance and biofilms. We emphasize design elements and properties of nanomaterials that can be engineered to enhance potency. Lastly, we present recent progress and remaining challenges for widespread clinical implementation of nanomaterials as antimicrobial therapeutics.
Collapse
Affiliation(s)
| | - Ahmed Nabawy
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| | - Cheng-Hsuan Li
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| | - Suzannah Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA.
| |
Collapse
|
33
|
Loo YS, Bose RJ, McCarthy JR, Mat Azmi ID, Madheswaran T. Biomimetic bacterial and viral-based nanovesicles for drug delivery, theranostics, and vaccine applications. Drug Discov Today 2020; 26:902-915. [PMID: 33383213 DOI: 10.1016/j.drudis.2020.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 01/04/2023]
Abstract
Smart nanocarriers obtained from bacteria and viruses offer excellent biomimetic properties which has led to significant research into the creation of advanced biomimetic materials. Their versatile biomimicry has application as biosensors, biomedical scaffolds, immobilization, diagnostics, and targeted or personalized treatments. The inherent natural traits of biomimetic and bioinspired bacteria- and virus-derived nanovesicles show potential for their use in clinical vaccines and novel therapeutic drug delivery systems. The past few decades have seen significant progress in the bioengineering of bacteria and viruses to manipulate and enhance their therapeutic benefits. From a pharmaceutical perspective, biomimetics enable the safe integration of naturally occurring bacteria and virus particles to achieve high, stable rates of cellular transfection/infection and prolonged circulation times. In addition, biomimetic technologies can overcome safety concerns associated with live-attenuated and inactivated whole bacteria or viruses. In this review, we provide an update on the utilization of bacterial and viral particles as drug delivery systems, theranostic carriers, and vaccine/immunomodulation modalities.
Collapse
Affiliation(s)
- Yan Shan Loo
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia
| | - Rajendran Jc Bose
- Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY 13501, USA
| | - Jason R McCarthy
- Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY 13501, USA
| | - Intan Diana Mat Azmi
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia.
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil, 57000 Kuala Lumpur, Malaysia.
| |
Collapse
|
34
|
Guyon L, Groo AC, Malzert-Fréon A. Relevant Physicochemical Methods to Functionalize, Purify, and Characterize Surface-Decorated Lipid-Based Nanocarriers. Mol Pharm 2020; 18:44-64. [PMID: 33244972 DOI: 10.1021/acs.molpharmaceut.0c00857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Surface functionalization of lipid-based nanocarriers (LBNCs) with targeting ligands has attracted huge interest in the field of nanomedicines for their ability to overcome some physiological barriers and their potential to deliver an active molecule to a specific target without causing damage to healthy tissues. The principal objective of this review is to summarize the present knowledge on LBNC decoration used for biomedical applications, with an emphasis on the ligands used, the functionalization approaches, and the purification methods after ligand corona formation. The most potent experimental techniques for the LBNC surface characterization are described. The potential of promising methods such as nuclear magnetic resonance spectroscopy and isothermal titration calorimetry to characterize ligand surface corona is also outlined.
Collapse
Affiliation(s)
- Léna Guyon
- CERMN, UNICAEN Université de Caen Normandie, F-14000 Caen, France
| | - Anne-Claire Groo
- CERMN, UNICAEN Université de Caen Normandie, F-14000 Caen, France
| | | |
Collapse
|
35
|
Li Y, Liu Y, Ren Y, Su L, Li A, An Y, Rotello V, Zhang Z, Wang Y, Liu Y, Liu S, Liu J, Laman JD, Shi L, van der Mei HC, Busscher HJ. Coating of a Novel Antimicrobial Nanoparticle with a Macrophage Membrane for the Selective Entry into Infected Macrophages and Killing of Intracellular Staphylococci. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2004942. [PMID: 34737689 PMCID: PMC8562776 DOI: 10.1002/adfm.202004942] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Indexed: 05/22/2023]
Abstract
Internalization of Staphylococcus aureus by macrophages can inactivate bacterial killing mechanisms, allowing intracellular residence and dissemination of infection. Concurrently, these staphylococci can evade antibiotics that are frequently unable to pass mammalian cell membranes. A binary, amphiphilic conjugate composed of triclosan and ciprofloxacin is synthesized that self-assemble through micelle formation into antimicrobial nanoparticles (ANPs). These novel ANPs are stabilized through encapsulation in macrophage membranes, providing membrane-encapsulated, antimicrobial-conjugated NPs (Me-ANPs) with similar protein activity, Toll-like receptor expression and negative surface charge as their precursor murine macrophage/human monocyte cell lines. The combination of Toll-like receptors and negative surface charge allows uptake of Me-ANPs by infected macrophages/monocytes through positively charged, lysozyme-rich membrane scars created during staphylococcal engulfment. Me-ANPs are not engulfed by more negatively charged sterile cells possessing less lysozyme at their surface. The Me-ANPs kill staphylococci internalized in macrophages in vitro. Me-ANPs likewise kill staphylococci more effectively than ANPs without membrane-encapsulation or clinically used ciprofloxacin in a mouse peritoneal infection model. Similarly, organ infections in mice created by dissemination of infected macrophages through circulation in the blood are better eradicated by Me-ANPs than by ciprofloxacin. These unique antimicrobial properties of macrophage-monocyte Me-ANPs provide a promising direction for human clinical application to combat persistent infections.
Collapse
Affiliation(s)
- Yuanfeng Li
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Yong Liu
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Yijin Ren
- Department of Orthodontics, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Linzhu Su
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
| | - Ang Li
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
| | - Yingli An
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
| | - Vincent Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Zhanzhan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
| | - Yin Wang
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
| | - Sidi Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Jian Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, P. R. China
| | - Jon D Laman
- Department of Biomedical Sciences of Cells and Systems, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Materials and Ministry, Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P. R. China
| | - Henny C van der Mei
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Henk J Busscher
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
36
|
Ho DK, Christmann R, Murgia X, De Rossi C, Frisch S, Koch M, Schaefer UF, Loretz B, Desmaele D, Couvreur P, Lehr CM. Synthesis and Biopharmaceutical Characterization of Amphiphilic Squalenyl Derivative Based Versatile Drug Delivery Platform. Front Chem 2020; 8:584242. [PMID: 33195079 PMCID: PMC7604382 DOI: 10.3389/fchem.2020.584242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Limited drug loading capacity (LC), mostly below 5% w/w, is a significant drawback of nanoparticulate drug delivery systems (DDS). Squalenoylation technology, which employs bioconjugation of squalenyl moiety and drug, allows self-assemble of nanoparticles (NPs) in aqueous media with significantly high LC (>30% w/w). The synthesis and particle preparation of squalenoylated prodrugs are, however, not facile for molecules with multiple reactive groups. Taking a different approach, we describe the synthesis of amphiphilic squalenyl derivatives (SqDs) as well as the physicochemical and biopharmaceutical characterizations of their self-assembled NPs as DDSs. The SqDs included in this study are (i) cationic squalenyl diethanolamine (ii) PEGylated SqD (PEG 750 Da), (iii) PEGylated SqD (PEG 3,000 Da), and (iv) anionic squalenyl hydrogen sulfate. All four SqDs self-assemble into NPs in a size range from 100 to 200 nm in an aqueous solution. Furthermore, all NP derivatives demonstrate appropriate biocompatibility and adequate colloidal stability in physiological relevant pH environments. The mucoprotein binding of PEGylated NPs is reduced compared to the charged NPs. Most importantly, this technology allows excellent LC (at maximum of 45% w/w) of a wide range of multifunctional compounds, varying in physicochemical properties and molecular weight. Interestingly, the drug release profile can be tuned by different loading methods. In summary, the SqD-based NPs appear as versatile drug delivery platforms.
Collapse
Affiliation(s)
- Duy-Khiet Ho
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Rebekka Christmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Xabier Murgia
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Chiara De Rossi
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany
| | - Sarah Frisch
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Marcus Koch
- INM-Leibniz Institute for New Materials, Saarbrücken, Germany
| | | | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany
| | - Didier Desmaele
- Faculté de Pharmacie, Institut Galien Paris Sud, Université Paris-Saclay, Chatenay-Malabry, France
| | - Patrick Couvreur
- Faculté de Pharmacie, Institut Galien Paris Sud, Université Paris-Saclay, Chatenay-Malabry, France
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
37
|
Youhanna S, Lauschke VM. The Past, Present and Future of Intestinal In Vitro Cell Systems for Drug Absorption Studies. J Pharm Sci 2020; 110:50-65. [PMID: 32628951 DOI: 10.1016/j.xphs.2020.07.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022]
Abstract
The intestinal epithelium acts as a selective barrier for the absorption of water, nutrients and orally administered drugs. To evaluate the gastrointestinal permeability of a candidate molecule, scientists and drug developers have a multitude of cell culture models at their disposal. Static transwell cultures constitute the most extensively characterized intestinal in vitro system and can accurately categorize molecules into low, intermediate and high permeability compounds. However, they lack key aspects of intestinal physiology, including the cellular complexity of the intestinal epithelium, flow, mechanical strain, or interactions with intestinal mucus and microbes. To emulate these features, a variety of different culture paradigms, including microfluidic chips, organoids and intestinal slice cultures have been developed. Here, we provide an updated overview of intestinal in vitro cell culture systems and critically review their suitability for drug absorption studies. The available data show that these advanced culture models offer impressive possibilities for emulating intestinal complexity. However, there is a paucity of systematic absorption studies and benchmarking data and it remains unclear whether the increase in model complexity and costs translates into improved drug permeability predictions. In the absence of such data, conventional static transwell cultures remain the current gold-standard paradigm for drug absorption studies.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
38
|
Marchianò V, Matos M, Serrano-Pertierra E, Gutiérrez G, Blanco-López MC. Vesicles as antibiotic carrier: State of art. Int J Pharm 2020; 585:119478. [PMID: 32473370 DOI: 10.1016/j.ijpharm.2020.119478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/15/2020] [Accepted: 05/24/2020] [Indexed: 12/22/2022]
Abstract
Antimicrobial resistance (AMR) has become a global health problem. Bacteria are able to adapt to different environments, with the presence or absence of a host, forming colonies and biofilms. In fact, biofilm formation confers chemical protection to the microbial cells, thus making most of the conventional antibiotics ineffective. Prevention and destruction of biofilms is a challenging task that should be addressed by a multidisciplinary approach from different research fields. One of the medical strategies used against biofilms is the therapy with drug delivery systems. Lipidic nanovesicles are a good choice for encapsulating drugs, increasing their pharmacodynamics and reducing side effects. These soft nanovesicles show significant advantages for their high biocompatibility, physical and chemistry properties, good affinity with drugs, and easy route of administration. This review summarizes the current knowledge on different types of vesicles which may be used as antibiotic carriers. The main preparation and purification methods for the synthesis of these vesicles are also presented. The advantages of drug encapsulation are critically reviewed. In addition, recent works on endolysin formulations as novel, "greener" and efficient antibiofilm solution are included. This paper can provide useful background for the design of novel efficient formulations and synergistic nanomaterials and could be also useful at the pharmaceutical industry to develop wastewater treatments and reduce the antibiotics in the environmental waters.
Collapse
Affiliation(s)
- Verdiana Marchianò
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain; Department of Chemical Engineering and Environmental Technology, University of Oviedo, 33006, Spain
| | - María Matos
- Department of Chemical Engineering and Environmental Technology, University of Oviedo, 33006, Spain; Institute of Biotechnology of Asturias, University of Oviedo, 33006, Spain
| | - Esther Serrano-Pertierra
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain; Institute of Biotechnology of Asturias, University of Oviedo, 33006, Spain
| | - Gemma Gutiérrez
- Department of Chemical Engineering and Environmental Technology, University of Oviedo, 33006, Spain; Institute of Biotechnology of Asturias, University of Oviedo, 33006, Spain.
| | - M C Blanco-López
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain; Institute of Biotechnology of Asturias, University of Oviedo, 33006, Spain.
| |
Collapse
|
39
|
Dubashynskaya NV, Skorik YA. Polymyxin Delivery Systems: Recent Advances and Challenges. Pharmaceuticals (Basel) 2020; 13:E83. [PMID: 32365637 PMCID: PMC7281078 DOI: 10.3390/ph13050083] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Polymyxins are vital antibiotics for the treatment of multiresistant Gram-negative ESKAPE pathogen infections. However, their clinical value is limited by their high nephrotoxicity and neurotoxicity, as well as their poor permeability and absorption in the gastrointestinal tract. This review focuses on various polymyxin delivery systems that improve polymyxin bioavailability and reduce drug toxicity through targeted and controlled release. Currently, the most suitable systems for improving oral, inhalation, and parenteral polymyxin delivery are polymer particles, liposomes, and conjugates, while gels, polymer fibers, and membranes are attractive materials for topical administration of polymyxin for the treatment of infected wounds and burns. In general, the application of these systems protects polymyxin molecules from the negative effects of both physiological and pathological factors while achieving higher concentrations at the target site and reducing dosage and toxicity. Improving the properties of polymyxin will be of great interest to researchers who are focused on developing antimicrobial drugs that show increased efficacy and safety.
Collapse
Affiliation(s)
| | - Yury A. Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, St. Petersburg 199004, Russia;
| |
Collapse
|
40
|
Goes A, Lapuhs P, Kuhn T, Schulz E, Richter R, Panter F, Dahlem C, Koch M, Garcia R, Kiemer AK, Müller R, Fuhrmann G. Myxobacteria-Derived Outer Membrane Vesicles: Potential Applicability Against Intracellular Infections. Cells 2020; 9:cells9010194. [PMID: 31940898 PMCID: PMC7017139 DOI: 10.3390/cells9010194] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
In 2019, it was estimated that 2.5 million people die from lower tract respiratory infections annually. One of the main causes of these infections is Staphylococcus aureus, a bacterium that can invade and survive within mammalian cells. S. aureus intracellular infections are difficult to treat because several classes of antibiotics are unable to permeate through the cell wall and reach the pathogen. This condition increases the need for new therapeutic avenues, able to deliver antibiotics efficiently. In this work, we obtained outer membrane vesicles (OMVs) derived from the myxobacteria Cystobacter velatus strain Cbv34 and Cystobacter ferrugineus strain Cbfe23, that are naturally antimicrobial, to target intracellular infections, and investigated how they can affect the viability of epithelial and macrophage cell lines. We evaluated by cytometric bead array whether they induce the expression of proinflammatory cytokines in blood immune cells. Using confocal laser scanning microscopy and flow cytometry, we also investigated their interaction and uptake into mammalian cells. Finally, we studied the effect of OMVs on planktonic and intracellular S. aureus. We found that while Cbv34 OMVs were not cytotoxic to cells at any concentration tested, Cbfe23 OMVs affected the viability of macrophages, leading to a 50% decrease at a concentration of 125,000 OMVs/cell. We observed only little to moderate stimulation of release of TNF-alpha, IL-8, IL-6 and IL-1beta by both OMVs. Cbfe23 OMVs have better interaction with the cells than Cbv34 OMVs, being taken up faster by them, but both seem to remain mostly on the cell surface after 24 h of incubation. This, however, did not impair their bacteriostatic activity against intracellular S. aureus. In this study, we provide an important basis for implementing OMVs in the treatment of intracellular infections.
Collapse
Affiliation(s)
- Adriely Goes
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Philipp Lapuhs
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Thomas Kuhn
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Eilien Schulz
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
| | - Robert Richter
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Helmholtz Centre for Infection Research (HZI), Department of Drug Delivery (DDEL), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany
| | - Fabian Panter
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
| | - Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Marcus Koch
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany;
| | - Ronald Garcia
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
| | - Alexandra K. Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66123 Saarbrücken, Germany; (C.D.); (A.K.K.)
| | - Rolf Müller
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Helmholtz Centre for Infection Research (HZI), Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (F.P.); (R.G.)
- German Center for Infection Research (DZIF), 38124 Braunschweig, Germany
| | - Gregor Fuhrmann
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany; (A.G.); (P.L.); (T.K.); (E.S.)
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany; (R.R.); (R.M.)
- Correspondence: ; Tel.: +49-68-198-806 (ext. 1500)
| |
Collapse
|
41
|
Schulz E, Karagianni A, Koch M, Fuhrmann G. Hot EVs - How temperature affects extracellular vesicles. Eur J Pharm Biopharm 2019; 146:55-63. [PMID: 31805356 DOI: 10.1016/j.ejpb.2019.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/15/2019] [Accepted: 11/27/2019] [Indexed: 12/25/2022]
Abstract
In recent years, extracellular vesicles (EVs) and outer membrane vesicles (OMVs) have become an extensive and diverse field of research. They hold potential as diagnostic markers, therapeutics and for fundamental biological understanding. Despite ongoing studies, numerous information regarding function, content and stability of EVs remains unclear. If EVs and OMVs ought to be used as therapeutics and in clinical environments, their stability is one of the most important factors to be considered. Especially for formulation development, EVs and OMVs need to be stable at higher temperatures. To the best of our knowledge, very little work has been published regarding heat stability of neither EVs nor OMVs. In the present study, we investigated B lymphoblastoid cell-derived EVs and OMVs derived from myxobacterial species Sorangiineae as model vesicles. We exposed the vesicles to 37 °C, 50 °C, 70 °C and 100 °C for 1 h, 6 h and 24 h, and also autoclaved them. Interestingly, physico-chemical analyses such as size, particle concentration and protein concentration showed minor alterations, particularly at 37 °C. Flow cytometry analysis emphasised these results suggesting that after heat impact, EVs and OMVs were still able to be taken up by macrophage-like dTHP-1 cells. These data indicate that both mammalian and bacterial vesicles show intrinsic stability at physiological temperature. Our findings are important to consider for vesicle formulation and for advanced bioengineering approaches.
Collapse
Affiliation(s)
- Eilien Schulz
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken 66123, Germany
| | - Anna Karagianni
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2.2, Saarbrücken 66123, Germany
| | - Gregor Fuhrmann
- Biogenic Nanotherapeutics Group (BION), Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, Saarbrücken 66123, Germany; Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken 66123, Germany.
| |
Collapse
|