1
|
Xing J, Han R, Zhao J, Zhang Y, Zhang M, Zhang Y, Zhang H, Nang SC, Zhai Y, Yuan L, Wang S, Wu H. Revisiting therapeutic options against resistant klebsiella pneumoniae infection: Phage therapy is key. Microbiol Res 2025; 293:128083. [PMID: 39904002 DOI: 10.1016/j.micres.2025.128083] [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: 10/01/2024] [Revised: 01/10/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Multi-drug resistant and carbapenem-resistant hypervirulent Klebsiella pneumoniae strains are spreading globally at an alarming rate, emerging as one of the most serious threats to global public health. The formidable challenges posed by the current arsenal of antimicrobials highlight the urgent need for novel strategies to combat K. pneumoniae infections. This review begins with a comprehensive analysis of the global dissemination of virulence factors and critical resistance profiles in K. pneumoniae, followed by an evaluation of the accessibility of novel therapeutic approaches for treating K. pneumoniae in clinical settings. Among these, phage therapy stands out for its considerable potential in addressing life-threatening K. pneumoniae infections. We critically examine the existing preclinical and clinical evidence supporting phage therapy, identifying key limitations that impede its broader clinical adoption. Additionally, we rigorously explore the role of genetic engineering in expanding the host range of K. pneumoniae phages, and discuss the future trajectory of this technology. In light of the 'Bad Bugs, No Drugs' era, we advocate leveraging artificial intelligence and deep learning to optimize and expand the application of phage therapy, representing a crucial advancement in the fight against the escalating threat of K. pneumoniae infections.
Collapse
Affiliation(s)
- Jiabao Xing
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Rongjia Han
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jinxin Zhao
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Yuying Zhang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Meng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yichao Zhang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Hang Zhang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Sue C Nang
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Yajun Zhai
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Li Yuan
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Shanmei Wang
- Department of Microbiology Laboratory, Henan Provincial People's Hospital, Zhengzhou, China.
| | - Hua Wu
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China; Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
2
|
Kumari R, Saraogi I. Navigating Antibiotic Resistance in Gram-Negative Bacteria: Current Challenges and Emerging Therapeutic Strategies. Chemphyschem 2025:e202401057. [PMID: 39970066 DOI: 10.1002/cphc.202401057] [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: 11/19/2024] [Revised: 01/30/2025] [Indexed: 02/21/2025]
Abstract
The rapid rise of antibiotic resistance poses a severe global health crisis, necessitating new approaches to counter this growing threat. The problem is exacerbated in Gram-negative bacterial pathogens as many antibiotics are unable to enter these cells owing to their unique additional outer membrane barrier. In this review, we discuss the challenges of targeting Gram-negative bacteria, including the complexity of the outer membrane, as well as the presence of efflux pumps and β-lactamases that contribute to resistance. We also review solutions proposed to facilitate the entry and accumulation of antibiotics in Gram-negative bacteria. These involve using existing antibiotics in combination with other inhibitors to attack the bacterial cell synergistically. We also highlight approaches to target Gram-negative pathogens via novel modes of action, providing new strategies to tackle antibiotic resistance.
Collapse
Affiliation(s)
- Reshma Kumari
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| | - Ishu Saraogi
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| |
Collapse
|
3
|
Siekierska I, Burmistrz M, Trylska J. Evaluating delivery of peptide nucleic acids to Gram-negative bacteria using differently linked membrane-active peptides and their stapled analogs. Bioorg Med Chem Lett 2024; 114:129993. [PMID: 39426432 DOI: 10.1016/j.bmcl.2024.129993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/27/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Antisense oligonucleotides have been developed as therapeutic compounds, with peptide nucleic acid (PNA) emerging as a promising nucleic acid mimic for antimicrobial applications. To be effective, PNAs must be internalized into bacterial cells, as they are not naturally absorbed. A strategy to improve PNA membrane penetration and cellular uptake involves covalently conjugating them to cell-penetrating peptides. However, these membrane-active peptides can exhibit cytotoxicity, and their efficiency as PNA carriers needs to be enhanced. Therefore, we explored new peptide-PNA conjugates and their linkers to understand how they affect PNA uptake into bacteria. We conjugated PNA to two peptides, anoplin and (KFF)3K, along with their structurally stabilized hydrocarbon-stapled derivatives, and evaluated their transport into various bacterial strains. The PNA sequence targeted bacterial mRNA encoding the essential acyl carrier protein. As linkages, we used either a non-cleavable 8-amino-2,6-dioxaoctanoyl (ethylene glycol, eg1) linker or a reducible disulfide bridge. We found that the hydrocarbon-stapled peptides did not enhance PNA delivery, despite the strong inner- and outer-membrane-penetrating capabilities of the standalone peptides. Additionally, the disulfide bridge linkage, which is cleavable in the bacterial cytoplasm, decreased the antimicrobial activity of the peptide-PNA conjugates. Notably, we identified anoplin as a new potent PNA carrier peptide, with the anoplin-eg1-PNA conjugate demonstrating antibacterial activity against E. coli and S. Typhimurium strains in the 2-4 µM range.
Collapse
Affiliation(s)
- Izabela Siekierska
- Centre of New Technologies, University of Warsaw, Banacha 2C, 02-097 Warsaw, Poland
| | - Michał Burmistrz
- Centre of New Technologies, University of Warsaw, Banacha 2C, 02-097 Warsaw, Poland
| | - Joanna Trylska
- Centre of New Technologies, University of Warsaw, Banacha 2C, 02-097 Warsaw, Poland.
| |
Collapse
|
4
|
Pals MJ, Lindberg A, Velema WA. Chemical strategies for antisense antibiotics. Chem Soc Rev 2024; 53:11303-11320. [PMID: 39436264 PMCID: PMC11495246 DOI: 10.1039/d4cs00238e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Indexed: 10/23/2024]
Abstract
Antibacterial resistance is a severe threat to modern medicine and human health. To stay ahead of constantly-evolving bacteria we need to expand our arsenal of effective antibiotics. As such, antisense therapy is an attractive approach. The programmability allows to in principle target any RNA sequence within bacteria, enabling tremendous selectivity. In this Tutorial Review we provide guidelines for devising effective antibacterial antisense agents and offer a concise perspective for future research. We will review the chemical architectures of antibacterial antisense agents with a special focus on the delivery and target selection for successful antisense design. This Tutorial Review will strive to serve as an essential guide for antibacterial antisense technology development.
Collapse
Affiliation(s)
- Mathijs J Pals
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Alexander Lindberg
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Willem A Velema
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| |
Collapse
|
5
|
Zhu J, Liang Z, Yao H, Wu Z. Identifying Cell-Penetrating Peptides for Effectively Delivering Antimicrobial Molecules into Streptococcus suis. Antibiotics (Basel) 2024; 13:725. [PMID: 39200025 PMCID: PMC11350675 DOI: 10.3390/antibiotics13080725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Cell-penetrating peptides (CPPs) are promising carriers to effectively transport antisense oligonucleotides (ASOs), including peptide nucleic acids (PNAs), into bacterial cells to combat multidrug-resistant bacterial infections, demonstrating significant therapeutic potential. Streptococcus suis, a Gram-positive bacterium, is a major bacterial pathogen in pigs and an emerging zoonotic pathogen. In this study, through the combination of super-resolution structured illumination microscopy (SR-SIM), flow cytometry analysis, and toxicity analysis assays, we investigated the suitability of four CPPs for delivering PNAs into S. suis cells: HIV-1 TAT efficiently penetrated S. suis cells with low toxicity against S. suis; (RXR)4XB had high penetration efficiency with inherent toxicity against S. suis; (KFF)3K showed lower penetration efficiency than HIV-1 TAT and (RXR)4XB; K8 failed to penetrate S. suis cells. HIV-1 TAT-conjugated PNA specific for the essential gyrase A subunit gene (TAT-anti-gyrA PNA) effectively inhibited the growth of S. suis. TAT-anti-gyrA PNA exhibited a significant bactericidal effect on serotypes 2, 4, 5, 7, and 9 strains of S. suis, which are known to cause human infections. Our study demonstrates the potential of CPP-ASO conjugates as new antimicrobial compounds for combating S. suis infections. Furthermore, our findings demonstrate that applying SR-SIM and flow cytometry analysis provides a convenient, intuitive, and cost-effective approach to identifying suitable CPPs for delivering cargo molecules into bacterial cells.
Collapse
Affiliation(s)
- Jinlu Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Zijing Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; (J.Z.); (Z.L.); (H.Y.)
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
- Guangdong Provincial Key Laboratory of Research on the Technology of Pig-Breeding and Pig-Disease Prevention, Guangzhou 511400, China
| |
Collapse
|
6
|
Pals MJ, Wijnberg L, Yildiz Ç, Velema WA. Catechol-Siderophore Mimics Convey Nucleic Acid Therapeutics into Bacteria. Angew Chem Int Ed Engl 2024; 63:e202402405. [PMID: 38407513 DOI: 10.1002/anie.202402405] [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: 02/04/2024] [Revised: 02/23/2024] [Accepted: 02/24/2024] [Indexed: 02/27/2024]
Abstract
Antibacterial resistance is a major threat for human health. There is a need for new antibacterials to stay ahead of constantly-evolving resistant bacteria. Nucleic acid therapeutics hold promise as powerful antibiotics, but issues with their delivery hamper their applicability. Here, we exploit the siderophore-mediated iron uptake pathway to efficiently transport antisense oligomers into bacteria. We appended a synthetic siderophore to antisense oligomers targeting the essential acpP gene in Escherichia coli. Siderophore-conjugated PNA and PMO antisense oligomers displayed potent antibacterial properties. Conjugates bearing a minimal siderophore consisting of a mono-catechol group showed equally effective. Targeting the lacZ transcript resulted in dose-dependent decreased β-galactosidase production, demonstrating selective protein downregulation. Applying this concept to Acinetobacter baumannii also showed concentration-dependent growth inhibition. Whole-genome sequencing of resistant mutants and competition experiments with the endogenous siderophore verified selective uptake through the siderophore-mediated iron uptake pathway. Lastly, no toxicity towards mammalian cells was found. Collectively, we demonstrate for the first time that large nucleic acid therapeutics can be efficiently transported into bacteria using synthetic siderophore mimics.
Collapse
Affiliation(s)
- Mathijs J Pals
- Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Luuk Wijnberg
- Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Çağlar Yildiz
- Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Willem A Velema
- Institute for Molecules and Materials, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| |
Collapse
|
7
|
Saini S, Goel K, Ghosh S, Das A, Saraogi I. Effects of PNA Sequence and Target Site Selection on Function of a 4.5S Non-Coding RNA. Chembiochem 2024:e202400029. [PMID: 38595046 DOI: 10.1002/cbic.202400029] [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: 01/12/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/11/2024]
Abstract
Peptide nucleic acid (PNA) based antisense strategy is a promising therapeutic approach to specifically inhibit target gene expression. However, unlike protein coding genes, identification of an ideal PNA binding site for non-coding RNA is not straightforward. Here, we compare the inhibitory activities of PNA molecules that bind a non-coding 4.5S RNA called SRP RNA, a key component of the bacterial signal recognition particle (SRP). A 9-mer PNA (PNA9) complementary to the tetraloop region of the RNA was more potent in inhibiting its interaction with the SRP protein, compared to an 8-mer PNA (PNA8) targeting a stem-loop. PNA9, which contained a homo-pyrimidine sequence could form a triplex with the complementary stretch of RNA in vitro as confirmed using a fluorescent derivative of PNA9 (F-PNA13). The RNA-PNA complex formation resulted in inhibition of SRP function with PNA9 and F-PNA13, but not PNA8 highlighting the importance of target site selection. Surprisingly, F-PNA13 which was more potent in inhibiting SRP function in vitro, showed weaker antibacterial activity compared to PNA9 likely due to poor cell penetration of the longer PNA. Our results underscore the importance of suitable target site selection and optimum PNA length to develop better antisense molecules against non-coding RNA.
Collapse
Affiliation(s)
- Snehlata Saini
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| | - Khushboo Goel
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| | - Sudipta Ghosh
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| | - Anirban Das
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| | - Ishu Saraogi
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, 462066, Madhya Pradesh, India
| |
Collapse
|
8
|
Moreira L, Guimarães NM, Santos RS, Loureiro JA, Pereira MC, Azevedo NF. Promising strategies employing nucleic acids as antimicrobial drugs. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102122. [PMID: 38333674 PMCID: PMC10850860 DOI: 10.1016/j.omtn.2024.102122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Antimicrobial resistance (AMR) is a growing concern because it causes microorganisms to develop resistance to drugs commonly used to treat infections. This results in increased difficulty in treating infections, leading to higher mortality rates and significant economic effects. Investing in new antimicrobial agents is, therefore, necessary to prevent and control AMR. Antimicrobial nucleic acids have arisen as potential key players in novel therapies for AMR infections. They have been designed to serve as antimicrobials and to act as adjuvants to conventional antibiotics or to inhibit virulent mechanisms. This new category of antimicrobial drugs consists of antisense oligonucleotides and oligomers, DNAzymes, and transcription factor decoys, differing in terms of structure, target molecules, and mechanisms of action. They are synthesized using nucleic acid analogs to enhance their resistance to nucleases. Because bacterial envelopes are generally impermeable to oligonucleotides, delivery into the cytoplasm typically requires the assistance of nanocarriers, which can affect their therapeutic potency. Given that numerous factors contribute to the success of these antimicrobial drugs, this review aims to provide a summary of the key advancements in the use of oligonucleotides for treating bacterial infections. Their mechanisms of action and the impact of factors such as nucleic acid design, target sequence, and nanocarriers on the antimicrobial potency are discussed.
Collapse
Affiliation(s)
- Luís Moreira
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Nuno M. Guimarães
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Rita S. Santos
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana A. Loureiro
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria C. Pereira
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Nuno F. Azevedo
- LEPABE–Laboratory for Process Engineering, Environment, Biotechnology, and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE–Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
9
|
Story S, Bhaduri S, Ganguly S, Dakarapu R, Wicks SL, Bhadra J, Kwange S, Arya DP. Understanding Antisense Oligonucleotide Efficiency in Inhibiting Prokaryotic Gene Expression. ACS Infect Dis 2024; 10:971-987. [PMID: 38385613 DOI: 10.1021/acsinfecdis.3c00645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Oligonucleotides offer a unique opportunity for sequence specific regulation of gene expression in bacteria. A fundamental question to address is the choice of oligonucleotide, given the large number of options available. Different modifications varying in RNA binding affinities and cellular uptake are available but no comprehensive comparisons have been performed. Herein, the efficiency of blocking expression of β-galactosidase (β-Gal) in E. coli was evaluated utilizing different antisense oligomers (ASOs). Fluorescein (FAM)-labeled oligomers were used to understand their differences in bacterial uptake. Flow cytometry analysis revealed significant differences in uptake, with high fluorescence seen in cells treated with FAM-labeled peptidic nucleic acid (PNA), phosphorodiamidate morpholino oligonucleotide (PMO) and phosphorothioate (PS) oligomers, and low fluorescence observed in cells treated with phosphodiester (PO) oligomers. Thermal denaturation (Tm) of oligomer:RNA duplexes and isothermal titration calorimetry (ITC) studies reveal that ASO binding to target RNA demonstrates a good correlation between Tm and Kd values. There was no correlation between Kd values and reduction of β-Gal activity in bacterial cells. However, cell-free translation assays demonstrated a direct relationship between Kd values and inhibition of gene expression by antisense oligomers, with tight binding oligomers such as LNA being the most efficient. Membrane active compounds such as polymyxin B and A22 further improved the cellular uptake of FAM-PNA and FAM-PS oligomers in wild-type E. coli cells. PNA and PMO were most effective in cellular uptake and reducing β-Gal activity as compared to oligomers with PS or those with PO linkages. Overall, cell uptake of the oligomers is shown as the key determinant in predicting their differences in bacterial antisense inhibition, and the RNA affinity is the key determinant in inhibition of gene expression in cell free systems.
Collapse
Affiliation(s)
- Sandra Story
- NUBAD, LLC, Greenville, South Carolina 29605, United States
| | | | - Sudakshina Ganguly
- Laboratory of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | | | - Sarah L Wicks
- NUBAD, LLC, Greenville, South Carolina 29605, United States
| | - Jhuma Bhadra
- Laboratory of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Simeon Kwange
- NUBAD, LLC, Greenville, South Carolina 29605, United States
| | - Dev P Arya
- Laboratory of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
- NUBAD, LLC, Greenville, South Carolina 29605, United States
| |
Collapse
|
10
|
Popova KB, Penchovsky R. General and Specific Cytotoxicity of Chimeric Antisense Oligonucleotides in Bacterial Cells and Human Cell Lines. Antibiotics (Basel) 2024; 13:122. [PMID: 38391508 PMCID: PMC10885958 DOI: 10.3390/antibiotics13020122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
In the last two decades, antisense oligonucleotide technology has emerged as a promising approach to tackling various healthcare issues and diseases, such as antimicrobial resistance, cancer, and neurodegenerative diseases. Despite the numerous improvements in the structure and modifications of the antisense oligonucleotides (ASOs), there are still specific problems with their clinical efficacy and preclinical cytotoxicity results. To better understand the effects of the ASOs in this paper, we conducted many MTT assays to assess the general and specific cytotoxicity of four new chimeric ASOs in bacterial cells and human cell lines. We demonstrate the absence of inhibitory activity in the human pathogenic bacteria Staphylococcus aureus by non-specific ASOs. The pVEC-ASO1 and pVEC-ASO2 are designed to have no specific targets in S. aureus. They have only partial hybridization to the guanylate kinase mRNA. The pVEC-ASO3 targets UBA2 mRNA, a hallmark cancer pathology in MYC-driven cancer, while pVEC-ASO4 has no complementary sequences. We discovered some cytotoxicity of the non-specific ASOs in healthy and cancer human cell lines. The results are compared with two other ASOs, targeting specific mRNA in cancer cells. All ASOs are delivered into the cell via the cell-penetrating oligopeptide pVEC, which is attached to them. We draw a good correlation between the thermodynamic stability of ASO/target RNA and the toxicity effect in human cell lines. The data obtained signify the importance of thorough bioinformatic analysis and high specificity in designing and developing novel ASOs for safer therapeutic agents in clinical practice.
Collapse
Affiliation(s)
- Katya B Popova
- Laboratory of Synthetic Biology and Bioinformatics, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria
- Institute for Nuclear Research and Nuclear Energy, Bulgarian Academy of Sciences, 1784 Sofia, Bulgaria
| | - Robert Penchovsky
- Laboratory of Synthetic Biology and Bioinformatics, Faculty of Biology, Sofia University "St. Kliment Ohridski", 8 Dragan Tzankov Blvd., 1164 Sofia, Bulgaria
| |
Collapse
|
11
|
Sparmann A, Vogel J. RNA-based medicine: from molecular mechanisms to therapy. EMBO J 2023; 42:e114760. [PMID: 37728251 PMCID: PMC10620767 DOI: 10.15252/embj.2023114760] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
RNA-based therapeutics have the potential to revolutionize the treatment and prevention of human diseases. While early research faced setbacks, it established the basis for breakthroughs in RNA-based drug design that culminated in the extraordinarily fast development of mRNA vaccines to combat the COVID-19 pandemic. We have now reached a pivotal moment where RNA medicines are poised to make a broad impact in the clinic. In this review, we present an overview of different RNA-based strategies to generate novel therapeutics, including antisense and RNAi-based mechanisms, mRNA-based approaches, and CRISPR-Cas-mediated genome editing. Using three rare genetic diseases as examples, we highlight the opportunities, but also the challenges to wide-ranging applications of this class of drugs.
Collapse
Affiliation(s)
- Anke Sparmann
- Helmholtz Institute for RNA‐based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI)WürzburgGermany
| | - Jörg Vogel
- Helmholtz Institute for RNA‐based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI)WürzburgGermany
- Institute of Molecular Infection Biology (IMIB)University of WürzburgWürzburgGermany
| |
Collapse
|
12
|
Bhowmik P, Modi B, Roy P, Chowdhury A. Strategies to combat Gram-negative bacterial resistance to conventional antibacterial drugs: a review. Osong Public Health Res Perspect 2023; 14:333-346. [PMID: 37920891 PMCID: PMC10626324 DOI: 10.24171/j.phrp.2022.0323] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 07/11/2023] [Accepted: 08/15/2023] [Indexed: 11/04/2023] Open
Abstract
The emergence of antimicrobial resistance raises the fear of untreatable diseases. Antimicrobial resistance is a multifaceted and dynamic phenomenon that is the cumulative result of different factors. While Gram-positive pathogens, such as methicillin-resistant Staphylococcus aureus and Clostridium difficile, were previously the most concerning issues in the field of public health, Gram-negative pathogens are now of prime importance. The World Health Organization's priority list of pathogens mostly includes multidrug-resistant Gram-negative organisms particularly carbapenem-resistant Enterobacterales, carbapenem-resistant Pseudomonas aeruginosa, and extensively drug-resistant Acinetobacter baumannii. The spread of Gram-negative bacterial resistance is a global issue, involving a variety of mechanisms. Several strategies have been proposed to control resistant Gram-negative bacteria, such as the development of antimicrobial auxiliary agents and research into chemical compounds with new modes of action. Another emerging trend is the development of naturally derived antibacterial compounds that aim for targets novel areas, including engineered bacteriophages, probiotics, metal-based antibacterial agents, odilorhabdins, quorum sensing inhibitors, and microbiome-modifying agents. This review focuses on the current status of alternative treatment regimens against multidrug-resistant Gram-negative bacteria, aiming to provide a snapshot of the situation and some information on the broader context.
Collapse
Affiliation(s)
- Priyanka Bhowmik
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Barkha Modi
- Department of Microbiology, Techno India University, Kolkata, India
| | - Parijat Roy
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Antarika Chowdhury
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| |
Collapse
|
13
|
Magaña AJ, Sklenicka J, Pinilla C, Giulianotti M, Chapagain P, Santos R, Ramirez MS, Tolmasky ME. Restoring susceptibility to aminoglycosides: identifying small molecule inhibitors of enzymatic inactivation. RSC Med Chem 2023; 14:1591-1602. [PMID: 37731693 PMCID: PMC10507813 DOI: 10.1039/d3md00226h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/21/2023] [Indexed: 09/22/2023] Open
Abstract
Growing resistance to antimicrobial medicines is a critical health problem that must be urgently addressed. Adding to the increasing number of patients that succumb to infections, there are other consequences to the rise in resistance like the compromise of several medical procedures and dental work that are heavily dependent on infection prevention. Since their introduction in the clinics, aminoglycoside antibiotics have been a critical component of the armamentarium to treat infections. Still, the increase in resistance and their side effects led to a decline in their utilization. However, numerous current factors, like the urgent need for antimicrobials and their favorable properties, led to renewed interest in these drugs. While efforts to design new classes of aminoglycosides refractory to resistance mechanisms and with fewer toxic effects are starting to yield new promising molecules, extending the useful life of those already in use is essential. For this, numerous research projects are underway to counter resistance from different angles, like inhibition of expression or activity of resistance components. This review focuses on selected examples of one aspect of this quest, the design or identification of small molecule inhibitors of resistance caused by enzymatic modification of the aminoglycoside. These compounds could be developed as aminoglycoside adjuvants to overcome resistant infections.
Collapse
Affiliation(s)
- Angel J Magaña
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Jan Sklenicka
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Clemencia Pinilla
- Center for Translational Science, Florida International University Port St. Lucie FL 34987 USA
| | - Marc Giulianotti
- Center for Translational Science, Florida International University Port St. Lucie FL 34987 USA
| | - Prem Chapagain
- Department of Physics, Florida International University Miami FL 33199 USA
- Biomolecular Sciences Institute, Florida International University Miami FL 33199 USA
| | - Radleigh Santos
- Department of Mathematics, Nova Southeastern University Fort Lauderdale FL 33314 USA
| | - Maria Soledad Ramirez
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Marcelo E Tolmasky
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| |
Collapse
|
14
|
Tekintaş Y, Temel A. Antisense oligonucleotides: a promising therapeutic option against infectious diseases. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 43:1-39. [PMID: 37395450 DOI: 10.1080/15257770.2023.2228841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
Infectious diseases have been one of the biggest health problems of humanity for centuries. Nucleic acid-based therapeutics have received attention in recent years with their effectiveness in the treatment of various infectious diseases and vaccine development studies. This review aims to provide a comprehensive understanding of the basic properties underlying the mechanism of antisense oligonucleotides (ASOs), their applications, and their challenges. The efficient delivery of ASOs is the greatest challenge for their therapeutic success, but this problem is overcome with new-generation antisense molecules developed with chemical modifications. The types, carrier molecules, and gene regions targeted by sequences have been summarized in detail. Research and development of antisense therapy is still in its infancy; however, gene silencing therapies appear to have the potential for faster and longer-lasting activity than conventional treatment strategies. On the other hand, realizing the potential of antisense therapy will require a large initial economic investment to ascertain the pharmacological properties and learn how to optimize them. The ability of ASOs to be rapidly designed and synthesized to target different microbes can reduce drug discovery time from 6 years to 1 year. Since ASOs are not particularly affected by resistance mechanisms, they come to the fore in the fight against antimicrobial resistance. The design-based flexibility of ASOs has enabled it to be used for different types of microorganisms/genes and successful in vitro and in vivo results have been revealed. The current review summarized a comprehensive understanding of ASO therapy in combating bacterial and viral infections.
Collapse
Affiliation(s)
- Yamaç Tekintaş
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Türkiye
| | - Aybala Temel
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Türkiye
| |
Collapse
|
15
|
Abstract
There is a continual debate on the pros and cons of broad-spectrum versus pathogen-specific antibiotics. The unmet need for a solution for antimicrobial resistance (AMR) has put this argument into sharper focus. A shortage of clinically differentiated antibiotics in late-stage clinical development coupled with the global unmet need in the face of the AMR onslaught has exacerbated the treatment options of drug-resistant bacterial infections. An added dimension to this problem is the current understanding of dysbiosis caused by antibiotics, often leading to negative fallout in immunocompromised patients. We attempt to deconstruct the nuances of this debate from an antibiotics discovery and a clinical standpoint.
Collapse
Affiliation(s)
- Santanu Datta
- Bugworks Research India Pvt Ltd, C-CAMP, NCBS, Bellary Road, Bangalore 560065, India
| |
Collapse
|
16
|
Fuchs M, Lamm-Schmidt V, Lenče T, Sulzer J, Bublitz A, Wackenreuter J, Gerovac M, Strowig T, Faber F. A network of small RNAs regulates sporulation initiation in Clostridioides difficile. EMBO J 2023:e112858. [PMID: 37140366 DOI: 10.15252/embj.2022112858] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 05/05/2023] Open
Abstract
The obligate anaerobic, enteric pathogen Clostridioides difficile persists in the intestinal tract by forming antibiotic-resistant endospores that contribute to relapsing and recurrent infections. Despite the importance of sporulation for C. difficile pathogenesis, environmental cues and molecular mechanisms that regulate sporulation initiation remain ill-defined. Here, by using RIL-seq to globally capture the Hfq-dependent RNA-RNA interactome, we discovered a network of small RNAs that bind to mRNAs encoding sporulation-related genes. We show that two of these small RNAs, SpoX and SpoY, regulate translation of the master regulator of sporulation, Spo0A, in an opposing manner, which ultimately leads to altered sporulation rates. Infection of antibiotic-treated mice with SpoX and SpoY deletion mutants revealed a global effect on gut colonization and intestinal sporulation. Our work uncovers an elaborate RNA-RNA interactome controlling the physiology and virulence of C. difficile and identifies a complex post-transcriptional layer in the regulation of spore formation in this important human pathogen.
Collapse
Affiliation(s)
- Manuela Fuchs
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for RNA-based Infection Research (HIRI), Würzburg, Germany
- Faculty of Medicine, Institute for Molecular Infection Biology (IMIB), Julius-Maximilians-University of Würzburg (JMU), Würzburg, Germany
| | - Vanessa Lamm-Schmidt
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for RNA-based Infection Research (HIRI), Würzburg, Germany
- Faculty of Medicine, Institute for Molecular Infection Biology (IMIB), Julius-Maximilians-University of Würzburg (JMU), Würzburg, Germany
| | - Tina Lenče
- Faculty of Medicine, Institute for Molecular Infection Biology (IMIB), Julius-Maximilians-University of Würzburg (JMU), Würzburg, Germany
| | - Johannes Sulzer
- Faculty of Medicine, Institute for Molecular Infection Biology (IMIB), Julius-Maximilians-University of Würzburg (JMU), Würzburg, Germany
| | - Arne Bublitz
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Janet Wackenreuter
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for RNA-based Infection Research (HIRI), Würzburg, Germany
| | - Milan Gerovac
- Faculty of Medicine, Institute for Molecular Infection Biology (IMIB), Julius-Maximilians-University of Würzburg (JMU), Würzburg, Germany
| | - Till Strowig
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig, Germany
| | - Franziska Faber
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for RNA-based Infection Research (HIRI), Würzburg, Germany
- Faculty of Medicine, Institute for Molecular Infection Biology (IMIB), Julius-Maximilians-University of Würzburg (JMU), Würzburg, Germany
| |
Collapse
|
17
|
Jung J, Popella L, Do PT, Pfau P, Vogel J, Barquist L. Design and off-target prediction for antisense oligomers targeting bacterial mRNAs with the MASON web server. RNA (NEW YORK, N.Y.) 2023; 29:570-583. [PMID: 36750372 PMCID: PMC10158992 DOI: 10.1261/rna.079263.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 01/10/2023] [Indexed: 05/06/2023]
Abstract
Antisense oligomers (ASOs), such as peptide nucleic acids (PNAs), designed to inhibit the translation of essential bacterial genes, have emerged as attractive sequence- and species-specific programmable RNA antibiotics. Yet, potential drawbacks include unwanted side effects caused by their binding to transcripts other than the intended target. To facilitate the design of PNAs with minimal off-target effects, we developed MASON (make antisense oligomers now), a web server for the design of PNAs that target bacterial mRNAs. MASON generates PNA sequences complementary to the translational start site of a bacterial gene of interest and reports critical sequence attributes and potential off-target sites. We based MASON's off-target predictions on experiments in which we treated Salmonella enterica serovar Typhimurium with a series of 10-mer PNAs derived from a PNA targeting the essential gene acpP but carrying two serial mismatches. Growth inhibition and RNA-sequencing (RNA-seq) data revealed that PNAs with terminal mismatches are still able to target acpP, suggesting wider off-target effects than anticipated. Comparison of these results to an RNA-seq data set from uropathogenic Escherichia coli (UPEC) treated with eleven different PNAs confirmed that our findings are not unique to Salmonella We believe that MASON's off-target assessment will improve the design of specific PNAs and other ASOs.
Collapse
Affiliation(s)
- Jakob Jung
- Institute for Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
| | - Linda Popella
- Institute for Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
| | - Phuong Thao Do
- Institute for Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), 97080 Würzburg, Germany
| | - Patrick Pfau
- Faculty of Medicine, University of Würzburg, 97080 Würzburg, Germany
| | - Jörg Vogel
- Institute for Molecular Infection Biology, University of Würzburg, 97080 Würzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), 97080 Würzburg, Germany
| | - Lars Barquist
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), 97080 Würzburg, Germany
- Faculty of Medicine, University of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
18
|
Araújo D, Gonçalves B, Vilas Boas D, Rodrigues ME, Henriques M, Silva S. Combined Application of Antisense Oligomers to Control Transcription Factors of Candida albicans Biofilm Formation. Mycopathologia 2023:10.1007/s11046-023-00734-0. [PMID: 37099227 DOI: 10.1007/s11046-023-00734-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/29/2023] [Indexed: 04/27/2023]
Abstract
Antisense oligomers (ASOs) have been little exploited to control determinants of Candida albicans virulence. Biofilm formation is an important virulence factor of C. albicans, that is regulated by a complex network of transcription factors (such as EFG1, BRG1 and ROB1). Thus, the main goal of this work was to project ASOs, based on the 2'-OMethyl chemical modification, to target BRG1 and ROB1 mRNA and to validate its application either alone or in combination with the EFG1 mRNA target, to reduce C. albicans biofilm formation. The ability of ASOs to control gene expression was evaluate by qRT-PCR. The effect on biofilm formation was determined by the total biomass quantification, and simultaneously the carbohydrates and proteins reduction on extracellular matrix. It was verified that all the oligomers were able to reduce the levels of gene expression and the ability of C. albicans to form biofilms. Furthermore, the combined application of the cocktail of ASOs enhances the inhibition of C. albicans biofilm formation, minimizing biofilm thickness by reducing the quantity of matrix content (protein and carbohydrate). So, our work confirms that ASOs are useful tools for research and therapeutic development on the control of Candida species biofilm formation.
Collapse
Affiliation(s)
- D Araújo
- Centre of Biological Engineering, LMaS-Laboratório de Microbiologia Aplicada à Saúde, CEB-Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal.
- LABBELS, Associate Laboratory, Braga/Guimarães, Portugal.
| | - B Gonçalves
- Centre of Biological Engineering, LMaS-Laboratório de Microbiologia Aplicada à Saúde, CEB-Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS, Associate Laboratory, Braga/Guimarães, Portugal
| | - D Vilas Boas
- Centre of Biological Engineering, LMaS-Laboratório de Microbiologia Aplicada à Saúde, CEB-Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS, Associate Laboratory, Braga/Guimarães, Portugal
| | - M E Rodrigues
- Centre of Biological Engineering, LMaS-Laboratório de Microbiologia Aplicada à Saúde, CEB-Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS, Associate Laboratory, Braga/Guimarães, Portugal
| | - M Henriques
- Centre of Biological Engineering, LMaS-Laboratório de Microbiologia Aplicada à Saúde, CEB-Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS, Associate Laboratory, Braga/Guimarães, Portugal
| | - S Silva
- Centre of Biological Engineering, LMaS-Laboratório de Microbiologia Aplicada à Saúde, CEB-Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- INIAV, IP-National Institute for Agrarian and Veterinary, Rua Dos Lagidos, Lugar da Madalena, Vairão, Vila Do Conde, Portugal
| |
Collapse
|
19
|
Alaoui Mdarhri H, Benmessaoud R, Yacoubi H, Seffar L, Guennouni Assimi H, Hamam M, Boussettine R, Filali-Ansari N, Lahlou FA, Diawara I, Ennaji MM, Kettani-Halabi M. Alternatives Therapeutic Approaches to Conventional Antibiotics: Advantages, Limitations and Potential Application in Medicine. Antibiotics (Basel) 2022; 11:1826. [PMID: 36551487 PMCID: PMC9774722 DOI: 10.3390/antibiotics11121826] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 12/23/2022] Open
Abstract
Resistance to antimicrobials and particularly multidrug resistance is one of the greatest challenges in the health system nowadays. The continual increase in the rates of antimicrobial resistance worldwide boosted by the ongoing COVID-19 pandemic poses a major public health threat. Different approaches have been employed to minimize the effect of resistance and control this threat, but the question still lingers as to their safety and efficiency. In this context, new anti-infectious approaches against multidrug resistance are being examined. Use of new antibiotics and their combination with new β-lactamase inhibitors, phage therapy, antimicrobial peptides, nanoparticles, and antisense antimicrobial therapeutics are considered as one such promising approach for overcoming bacterial resistance. In this review, we provide insights into these emerging alternative therapies that are currently being evaluated and which may be developed in the future to break the progression of antimicrobial resistance. We focus on their advantages and limitations and potential application in medicine. We further highlight the importance of the combination therapy approach, wherein two or more therapies are used in combination in order to more effectively combat infectious disease and increasing access to quality healthcare. These advances could give an alternate solution to overcome antimicrobial drug resistance. We eventually hope to provide useful information for clinicians who are seeking solutions to the problems caused by antimicrobial resistance.
Collapse
Affiliation(s)
- Hiba Alaoui Mdarhri
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Rachid Benmessaoud
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Houda Yacoubi
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Lina Seffar
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Houda Guennouni Assimi
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Mouhsine Hamam
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Rihabe Boussettine
- Laboratory of Virology, Oncology, Biosciences, Environment and New Energies, Faculty of Sciences and Techniques Mohammedia, University Hassan II of Casablanca, Casablanca 28 806, Morocco
| | - Najoie Filali-Ansari
- Laboratory of Virology, Oncology, Biosciences, Environment and New Energies, Faculty of Sciences and Techniques Mohammedia, University Hassan II of Casablanca, Casablanca 28 806, Morocco
| | - Fatima Azzahra Lahlou
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Idrissa Diawara
- Department of Biological Engineering, Higher Institute of Bioscience and Biotechnology, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| | - Moulay Mustapha Ennaji
- Laboratory of Virology, Oncology, Biosciences, Environment and New Energies, Faculty of Sciences and Techniques Mohammedia, University Hassan II of Casablanca, Casablanca 28 806, Morocco
| | - Mohamed Kettani-Halabi
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
- National Reference Laboratory, Mohammed VI University of Health Sciences (UM6SS), Casablanca 82 403, Morocco
| |
Collapse
|
20
|
Costa VG, Costa SM, Saramago M, Cunha MV, Arraiano CM, Viegas SC, Matos RG. Developing New Tools to Fight Human Pathogens: A Journey through the Advances in RNA Technologies. Microorganisms 2022; 10:2303. [PMID: 36422373 PMCID: PMC9697208 DOI: 10.3390/microorganisms10112303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 09/18/2024] Open
Abstract
A long scientific journey has led to prominent technological advances in the RNA field, and several new types of molecules have been discovered, from non-coding RNAs (ncRNAs) to riboswitches, small interfering RNAs (siRNAs) and CRISPR systems. Such findings, together with the recognition of the advantages of RNA in terms of its functional performance, have attracted the attention of synthetic biologists to create potent RNA-based tools for biotechnological and medical applications. In this review, we have gathered the knowledge on the connection between RNA metabolism and pathogenesis in Gram-positive and Gram-negative bacteria. We further discuss how RNA techniques have contributed to the building of this knowledge and the development of new tools in synthetic biology for the diagnosis and treatment of diseases caused by pathogenic microorganisms. Infectious diseases are still a world-leading cause of death and morbidity, and RNA-based therapeutics have arisen as an alternative way to achieve success. There are still obstacles to overcome in its application, but much progress has been made in a fast and effective manner, paving the way for the solid establishment of RNA-based therapies in the future.
Collapse
Affiliation(s)
| | | | | | | | | | - Sandra C. Viegas
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal; (V.G.C.); (S.M.C.); (M.S.); (M.V.C.); (C.M.A.)
| | - Rute G. Matos
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal; (V.G.C.); (S.M.C.); (M.S.); (M.V.C.); (C.M.A.)
| |
Collapse
|
21
|
Castro J, Barros MM, Araújo D, Campos AM, Oliveira R, Silva S, Almeida C. Swine enteric colibacillosis: Current treatment avenues and future directions. Front Vet Sci 2022; 9:981207. [PMID: 36387374 PMCID: PMC9650617 DOI: 10.3389/fvets.2022.981207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/10/2022] [Indexed: 09/10/2023] Open
Abstract
Enteric colibacillosis is a common disease in nursing and weanling pigs. It is caused by the colonization of the small intestine by enterotoxigenic strains of Escherichia coli (ETEC) that make use of specific fimbria or pili to adhere to the absorptive epithelial cells of the jejunum and ileum. Once attached, and when both the immunological systems and the gut microbiota are poorly developed, ETEC produce one or more enterotoxins that can have local and, further on, systemic effects. These enterotoxins cause fluid and electrolytes to be secreted into the intestinal lumen of animals, which results in diarrhea, dehydration, and acidosis. From the diversity of control strategies, antibiotics and zinc oxide are the ones that have contributed more significantly to mitigating post-weaning diarrhea (PWD) economic losses. However, concerns about antibiotic resistance determined the restriction on the use of critically important antimicrobials in food-producing animals and the prohibition of their use as growth promoters. As such, it is important now to begin the transition from these preventive/control measures to other, more sustainable, approaches. This review provides a quick synopsis of the currently approved and available therapies for PWD treatment while presenting an overview of novel antimicrobial strategies that are being explored for the control and treatment of this infection, including, prebiotics, probiotics, synbiotics, organic acids, bacteriophages, spray-dried plasma, antibodies, phytogenic substances, antisense oligonucleotides, and aptamers.
Collapse
Affiliation(s)
- Joana Castro
- National Institute for Agrarian and Veterinarian Research (INIAV), Vila do Conde, Portugal
| | - Maria Margarida Barros
- National Institute for Agrarian and Veterinarian Research (INIAV), Vila do Conde, Portugal
| | - Daniela Araújo
- National Institute for Agrarian and Veterinarian Research (INIAV), Vila do Conde, Portugal
| | - Ana Maria Campos
- National Institute for Agrarian and Veterinarian Research (INIAV), Vila do Conde, Portugal
| | - Ricardo Oliveira
- National Institute for Agrarian and Veterinarian Research (INIAV), Vila do Conde, Portugal
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Sónia Silva
- National Institute for Agrarian and Veterinarian Research (INIAV), Vila do Conde, Portugal
- Centre of Biological Engineering, Braga, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| | - Carina Almeida
- National Institute for Agrarian and Veterinarian Research (INIAV), Vila do Conde, Portugal
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
- Centre of Biological Engineering, Braga, Portugal
- LABBELS – Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
22
|
Rouse WB, Gart J, Peysakhova L, Moss WN. Analysis of key genes in Mycobacterium ulcerans reveals conserved RNA structural motifs and regions with apparent pressure to remain unstructured. FRONTIERS IN TROPICAL DISEASES 2022; 3. [PMID: 37006713 PMCID: PMC10062443 DOI: 10.3389/fitd.2022.1009362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Buruli Ulcer is a neglected tropical disease that results in disfiguring and dangerous lesions in affected persons across a wide geographic area, including much of West Africa. The causative agent of Buruli Ulcer is Mycobacterium ulcerans, a relative of the bacterium that causes tuberculosis and leprosy. Few therapeutic options exist for the treatment of this disease beyond antibiotics in the early stages, which are frequently ineffective, and surgical removal in the later stage. In this study we analyze six genes in Mycobacterium ulcerans that have high potential of therapeutic targeting. We focus our analysis on a combined in silico and comparative sequence study of potential RNA secondary structure across these genes. The result of this work was the comprehensive local RNA structural landscape across each of these significant genes. This revealed multiple sites of ordered and evolved RNA structure interspersed between sequences that either have no bias for structure or, indeed, appear to be ordered to be unstructured and (potentially) accessible. In addition to providing data that could be of interest to basic biology, our results provide guides for efforts aimed at targeting this pathogen at the RNA level. We explore this latter possibility through the in silico analysis of antisense oligonucleotides that could potentially be used to target pathogen RNA.
Collapse
Affiliation(s)
- Warren B. Rouse
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Jessica Gart
- Science and Engineering Research Program (SERP), Staten Island Technical High School, Staten Island, NY, United States
| | - Lauren Peysakhova
- Science and Engineering Research Program (SERP), Staten Island Technical High School, Staten Island, NY, United States
| | - Walter N. Moss
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
- CORRESPONDENCE: Walter N. Moss,
| |
Collapse
|
23
|
Wu S, Gan T, Xie L, Deng S, Liu Y, Zhang H, Hu X, Lei L. Antibacterial performance of graphene oxide/alginate-based antisense hydrogel for potential therapeutic application in Staphylococcus aureus infection. BIOMATERIALS ADVANCES 2022; 141:213121. [PMID: 36162343 DOI: 10.1016/j.bioadv.2022.213121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/29/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023]
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic bacterium that causes several infections in humans. However, chronic biofilms remain a major challenge associated with recalcitrance toward traditional treatments. Herein, an antibacterial hydrogel composed of antisense DNA oligonucleotides, graphene oxide and alginate is construed for biofilm management and infection care. The hydrogel is established through noncovalent binding and possesses injectability and degradability properties. Furthermore, hydrogels present controllable release of cargoes, genetic targeting antibacterial effects and stem cell supporting capabilities. Our in vivo results reveal a high antibiofilm performance and good biocompatibility, which significantly improve tissue regeneration. The hydrogel inhibits biofilm formation by decreasing the expression of YycFG with antisense and viability of strains by graphene oxide. Thus, antisense hydrogels can be a promising antibacterial bioactive material for potential therapeutic S. aureus infection.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tingjiang Gan
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liwei Xie
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shu Deng
- Boston University Henry M Goldman School of Dental Medicine, Boston, MA 02101, USA
| | - Yunjie Liu
- West China School of Public Health, Sichuan University, Chengdu City, Sichuan 610041, China
| | - Hui Zhang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuefeng Hu
- National Engineering Research Center for Biomaterials, Biomaterials Building, Sichuan University, 29 Wangjiang Road, Chengdu 610064, Sichuan, China.
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
24
|
Popella L, Jung J, Do PT, Hayward RJ, Barquist L, Vogel J. Comprehensive analysis of PNA-based antisense antibiotics targeting various essential genes in uropathogenic Escherichia coli. Nucleic Acids Res 2022; 50:6435-6452. [PMID: 35687096 PMCID: PMC9226493 DOI: 10.1093/nar/gkac362] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/05/2022] [Accepted: 06/08/2022] [Indexed: 12/13/2022] Open
Abstract
Antisense peptide nucleic acids (PNAs) that target mRNAs of essential bacterial genes exhibit specific bactericidal effects in several microbial species, but our mechanistic understanding of PNA activity and their target gene spectrum is limited. Here, we present a systematic analysis of PNAs targeting 11 essential genes with varying expression levels in uropathogenic Escherichia coli (UPEC). We demonstrate that UPEC is susceptible to killing by peptide-conjugated PNAs, especially when targeting the widely-used essential gene acpP. Our evaluation yields three additional promising target mRNAs for effective growth inhibition, i.e.dnaB, ftsZ and rpsH. The analysis also shows that transcript abundance does not predict target vulnerability and that PNA-mediated growth inhibition is not universally associated with target mRNA depletion. Global transcriptomic analyses further reveal PNA sequence-dependent but also -independent responses, including the induction of envelope stress response pathways. Importantly, we show that 9mer PNAs are generally as effective in inhibiting bacterial growth as their 10mer counterparts. Overall, our systematic comparison of a range of PNAs targeting mRNAs of different essential genes in UPEC suggests important features for PNA design, reveals a general bacterial response to PNA conjugates and establishes the feasibility of using PNA antibacterials to combat UPEC.
Collapse
Affiliation(s)
- Linda Popella
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080, Würzburg, Germany
| | - Jakob Jung
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080, Würzburg, Germany
| | - Phuong Thao Do
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), D-97080, Würzburg, Germany
| | - Regan J Hayward
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), D-97080, Würzburg, Germany
| | - Lars Barquist
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), D-97080, Würzburg, Germany
- Faculty of Medicine, University of Würzburg, D-97080, Würzburg, Germany
| | - Jörg Vogel
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080, Würzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), D-97080, Würzburg, Germany
- Faculty of Medicine, University of Würzburg, D-97080, Würzburg, Germany
| |
Collapse
|
25
|
Tian Y, Zhang Y, Zhang M, Chen X, Lei L, Hu T. Antisense vicR-Loaded Dendritic Mesoporous Silica Nanoparticles Regulate the Biofilm Organization and Cariogenicity of Streptococcus mutans. Int J Nanomedicine 2022; 17:1255-1272. [PMID: 35340824 PMCID: PMC8956320 DOI: 10.2147/ijn.s334785] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Purpose VicR is the essential response regulator related to the synthesis of exopolysaccharide (EPS) – one of the main cariogenic factors of S. mutans. An antisense vicR RNA (ASvicR) could bind to vicR mRNA, hindering the transcription and translation of the vicR gene. We had constructed a recombinant plasmid containing the ASvicR sequence (plasmid-ASvicR) and proved that it could reduce EPS synthesis, biofilm formation, and cariogenicity. However, the recombinant plasmids are supposed to be protected from enzymatic degradation and possess higher transformation efficiency. The principal objective of the present research was to construct an appropriate vector that can carry and protect the plasmid-ASvicR and investigate the effects of the carried plasmids on the cariogenicity of the S. mutans. Methods Aminated dendritic mesoporous silica nanoparticles (DMSNs-NH2) were synthesized and characterized. The ability of DMSNs-NH2 to carry and preserve the plasmid-ASvicR (DMSNs-NH2-ASvicR) was proved by the loading curve, agarose electrophoresis, DNase I digestion assays, and energy-dispersive spectrometry (EDS) mapping. Transformation assays demonstrated whether the plasmid could enter S. mutans. The effect of DMSNs-NH2-ASvicR on the 12-hour and 24-hour biofilms of S. mutans was evaluated by biofilm formation experiments and quantitative reverse transcription polymerase chain reaction (qRT-PCR). The cytotoxicity of DMSNs-NH2-ASvicR was assessed by CCK-8 and live/dead staining assays. The regulation of DMSNs-NH2-ASvicR on the cariogenicity of S. mutans was also evaluated in vivo. Results DMSNs-NH2 could load approximately 92% of plasmid-ASvicR at a mass ratio of 80 and protect most of plasmid-ASvicR from degradation by DNase I. The plasmid-ASvicR loaded on DMSNs-NH2 could be transformed into S. mutans, which down-regulated the expression of the vicR gene, reducing EPS synthesis and biofilm organization of S. mutans. DMSNs-NH2-ASvicR exhibited favorable biocompatibility, laying a foundation for its subsequent biomedical application. In addition, DMSNs-NH2-ASvicR led to decreased caries in vivo. Conclusion DMSNs-NH2 is a suitable vector of plasmid-ASvicR, and DMSNs-NH2-ASvicR can inhibit biofilm formation, reducing the cariogenicity of S. mutans. These findings reveal that DMSNs-NH2-ASvicR is a promising agent for preventing and treating dental caries.
Collapse
Affiliation(s)
- Yuting Tian
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yue Zhang
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Mengjiao Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xianchun Chen
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, People's Republic of China
| | - Lei Lei
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Tao Hu
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| |
Collapse
|
26
|
Halloy F, Biscans A, Bujold KE, Debacker A, Hill AC, Lacroix A, Luige O, Strömberg R, Sundstrom L, Vogel J, Ghidini A. Innovative developments and emerging technologies in RNA therapeutics. RNA Biol 2022; 19:313-332. [PMID: 35188077 PMCID: PMC8865321 DOI: 10.1080/15476286.2022.2027150] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
RNA-based therapeutics are emerging as a powerful platform for the treatment of multiple diseases. Currently, the two main categories of nucleic acid therapeutics, antisense oligonucleotides and small interfering RNAs (siRNAs), achieve their therapeutic effect through either gene silencing, splicing modulation or microRNA binding, giving rise to versatile options to target pathogenic gene expression patterns. Moreover, ongoing research seeks to expand the scope of RNA-based drugs to include more complex nucleic acid templates, such as messenger RNA, as exemplified by the first approved mRNA-based vaccine in 2020. The increasing number of approved sequences and ongoing clinical trials has attracted considerable interest in the chemical development of oligonucleotides and nucleic acids as drugs, especially since the FDA approval of the first siRNA drug in 2018. As a result, a variety of innovative approaches is emerging, highlighting the potential of RNA as one of the most prominent therapeutic tools in the drug design and development pipeline. This review seeks to provide a comprehensive summary of current efforts in academia and industry aimed at fully realizing the potential of RNA-based therapeutics. Towards this, we introduce established and emerging RNA-based technologies, with a focus on their potential as biosensors and therapeutics. We then describe their mechanisms of action and their application in different disease contexts, along with the strengths and limitations of each strategy. Since the nucleic acid toolbox is rapidly expanding, we also introduce RNA minimal architectures, RNA/protein cleavers and viral RNA as promising modalities for new therapeutics and discuss future directions for the field.
Collapse
Affiliation(s)
- François Halloy
- Department of Paediatrics, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Annabelle Biscans
- Oligonucleotide Chemistry, Discovery Sciences, BioPharmaceuticals R&d, AstraZeneca, Gothenburg, Sweden
| | - Katherine E. Bujold
- Department of Chemistry & Chemical Biology, McMaster University, (Ontario), Canada
| | | | - Alyssa C. Hill
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Eth Zürich, Zürich, Switzerland
| | - Aurélie Lacroix
- Sixfold Bioscience, Translation & Innovation Hub, London, UK
| | - Olivia Luige
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden
| | - Roger Strömberg
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden
| | - Linda Sundstrom
- Mechanistic and Structural Biology, Discovery Sciences, BioPharmaceuticals R&d, AstraZeneca, Gothenburg, Sweden
| | - Jörg Vogel
- Helmholtz Institute for RNA-based Infection Research (Hiri), Helmholtz Center for Infection Research (Hzi), Würzburg, Germany
- RNA Biology Group, Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Alice Ghidini
- Mechanistic and Structural Biology, Discovery Sciences, BioPharmaceuticals R&d, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
27
|
Nguyen PV, Aubry C, Boudaoud N, Gaubert A, Langlois MH, Marchivie M, Gaudin K, Arpin C, Barthélémy P, Kauss T. Oligonucleotide Solid Nucleolipid Nanoparticles against Antibiotic Resistance of ESBL-Producing Bacteria. Pharmaceutics 2022; 14:299. [PMID: 35214036 PMCID: PMC8876242 DOI: 10.3390/pharmaceutics14020299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Antibiotic resistance has become a major issue in the global healthcare system, notably in the case of Gram-negative bacteria. Recent advances in technology with oligonucleotides have an enormous potential for tackling this problem, providing their efficient intrabacterial delivery. The current work aimed to apply this strategy by using a novel nanoformulation consisting of DOTAU, a nucleolipid carrier, in an attempt to simultaneously deliver antibiotic and anti-resistance oligonucleotides. Ceftriaxone, a third-generation cephalosporin, was formulated with DOTAU to form an ion pair, and was then nanoprecipitated. The obtained solid nanocapsules were characterized using FT-IR, XRD, HPLC, TEM and DLS techniques and further functionalized by the anti-resistance ONα sequence. To obtain an optimal anti-resistance activity and encapsulation yield, both the formulation protocol and the concentration of ONα were optimized. As a result, monodispersed negatively charged nanoparticles of CFX-DOTAU-ONα with a molar ratio of 10:24:1 were obtained. The minimum inhibitory concentration of these nanoparticles on the resistant Escherichia coli strain was significantly reduced (by 75%) in comparison with that of non-vectorized ONα. All aforementioned results reveal that our nanoformulation can be considered as an efficient and relevant strategy for oligonucleotide intrabacterial delivery in the fight against antibiotic resistance.
Collapse
Affiliation(s)
- Phuoc Vinh Nguyen
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| | - Clémentine Aubry
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| | - Narimane Boudaoud
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| | - Alexandra Gaubert
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| | - Marie-Hélène Langlois
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| | - Mathieu Marchivie
- UMR 5026, University of Bordeaux, CNRS, Bordeaux-INP, ICMCB, 87 Avenue du Dr Albert Schweitzer, CEDEX, 33608 Pessac, France;
| | - Karen Gaudin
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| | - Corinne Arpin
- MFP, CNRS 5234, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France;
| | - Philippe Barthélémy
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| | - Tina Kauss
- ARNA, Inserm U1212, CNRS 5320, University of Bordeaux, 146 rue Léo Saignat, CEDEX, 33076 Bordeaux, France; (P.V.N.); (C.A.); (N.B.); (A.G.); (M.-H.L.); (K.G.); (P.B.)
| |
Collapse
|
28
|
Roncarati D, Scarlato V, Vannini A. Targeting of Regulators as a Promising Approach in the Search for Novel Antimicrobial Agents. Microorganisms 2022; 10:microorganisms10010185. [PMID: 35056634 PMCID: PMC8777881 DOI: 10.3390/microorganisms10010185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Since the discovery of penicillin in the first half of the last century, antibiotics have become the pillars of modern medicine for fighting bacterial infections. However, pathogens resistant to antibiotic treatment have increased in recent decades, and efforts to discover new antibiotics have decreased. As a result, it is becoming increasingly difficult to treat bacterial infections successfully, and we look forward to more significant efforts from both governments and the scientific community to research new antibacterial drugs. This perspective article highlights the high potential of bacterial transcriptional and posttranscriptional regulators as targets for developing new drugs. We highlight some recent advances in the search for new compounds that inhibit their biological activity and, as such, appear very promising for treating bacterial infections.
Collapse
Affiliation(s)
- Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
- Correspondence: (D.R.); (V.S.); (A.V.)
| | - Vincenzo Scarlato
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
- Correspondence: (D.R.); (V.S.); (A.V.)
| | - Andrea Vannini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Correspondence: (D.R.); (V.S.); (A.V.)
| |
Collapse
|
29
|
OUP accepted manuscript. Med Mycol 2022; 60:6576775. [DOI: 10.1093/mmy/myac030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/11/2022] [Accepted: 04/29/2022] [Indexed: 11/14/2022] Open
|
30
|
Afridi M, Khan SA, Afridi R, Ullah F, Majid A, Khan AA, Ali N. Combining antibiotics with silver nanoparticles: A potential treatment strategy against antimicrobial resistance. MAIN GROUP CHEMISTRY 2021. [DOI: 10.3233/mgc-210131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Growing resistance to currently approved antibiotics is posing serious concern worldwide. The multidrug-resistant organisms are a major cause of mortality and morbidity around the globe. The limited options to treat infections caused by resistant organism requires alternative strategies to increase the effectiveness of antibiotic for better clinical outcomes. Recent advances in nanotechnology have enabled the drugs to be used in nanoscale to increase the effectiveness of antibiotics. The use of nanoparticles to treat infectious diseases has a long history in the pharmaceutical market, and the versatility of these particles to incorporate various materials as carriers make it an attractive option to combat the current crisis of emerging antibacterial resistance. Silver, a metal with many medical applications, has inherent antimicrobial properties. Therefore, silver NPs are appearing as one of the best options to be used in combination with antibiotics to increase effectiveness against resistant bacteria. Here, we discuss the applications and mechanisms of silver NPs to treat microbial resistance in light of recent research.
Collapse
Affiliation(s)
- Maryam Afridi
- Department of Pharmacy, Institute of Chemical and Pharmaceutical Sciences, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Saeed Ahmad Khan
- Department of Pharmacy, Institute of Chemical and Pharmaceutical Sciences, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Ruqayya Afridi
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Farman Ullah
- Department of Pharmacy, Institute of Chemical and Pharmaceutical Sciences, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Majid
- Department of Zoology, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa
| | - Aziz Ahmad Khan
- Department of Pharmacy, Institute of Chemical and Pharmaceutical Sciences, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Nawab Ali
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
31
|
Araújo D, Mil-Homens D, Rodrigues ME, Henriques M, Jørgensen PT, Wengel J, Silva S. Antisense locked nucleic acid gapmers to control Candida albicans filamentation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 39:102469. [PMID: 34606999 DOI: 10.1016/j.nano.2021.102469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022]
Abstract
Whereas locked nucleic acid (LNA) has been extensively used to control gene expression, it has never been exploited to control Candida virulence genes. Thus, the main goal of this work was to compare the efficacy of five different LNA-based antisense oligonucleotides (ASO) with respect to the ability to control EFG1 gene expression, to modulate filamentation and to reduce C. albicans virulence. In vitro, all LNA-ASOs were able to significantly reduce C. albicans filamentation and to control EFG1 gene expression. Using the in vivo Galleria mellonella model, important differences among the five LNA-ASOs were revealed in terms of C. albicans virulence reduction. The inclusion of PS-linkage and palmitoyl-2'-amino-LNA chemical modification in these five LNA gapmers proved to be the most promising combination, increasing the survival of G. mellonella by 40%. Our work confirms that LNA-ASOs are useful tools for research and therapeutic development in the candidiasis field.
Collapse
Affiliation(s)
- Daniela Araújo
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Dalila Mil-Homens
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Lisbon University, Lisbon, Portugal
| | - Maria Elisa Rodrigues
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Mariana Henriques
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Per Trolle Jørgensen
- Biomolecular Nanoscale Engineering Center, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Jesper Wengel
- Biomolecular Nanoscale Engineering Center, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Sónia Silva
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, Braga, Portugal; National Institute for Agrarian and Veterinary Research, Vairão, Vila do Conde, Portugal.
| |
Collapse
|
32
|
Henderson CA, Vincent HA, Callaghan AJ. Reprogramming Gene Expression by Targeting RNA-Based Interactions: A Novel Pipeline Utilizing RNA Array Technology. ACS Synth Biol 2021; 10:1847-1858. [PMID: 34283568 DOI: 10.1021/acssynbio.0c00603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Regulatory RNA-based interactions are critical for coordinating gene expression and are increasingly being targeted in synthetic biology, antimicrobial, and therapeutic fields. Bacterial trans-encoded small RNAs (sRNAs) regulate the translation and/or stability of mRNA targets through base-pairing interactions. These interactions are often integral to complex gene circuits which coordinate critical bacterial processes. The ability to predictably modulate these gene circuits has potential for reprogramming gene expression for synthetic biology and antibacterial purposes. Here, we present a novel pipeline for targeting such RNA-based interactions with antisense oligonucleotides (ASOs) in order to reprogram gene expression. As proof-of-concept, we selected sRNA-mRNA interactions that are central to the Vibrio cholerae quorum sensing pathway, required for V. cholerae pathogenesis, as a regulatory RNA-based interaction input. We rationally designed anti-sRNA ASOs to target the sRNAs and synthesized them as peptide nucleic acids (PNAs). Next, we devised an RNA array-based interaction assay to allow screening of the anti-sRNA ASOs in vitro. Finally, an Escherichia coli-based gene expression reporter assay was developed and used to validate anti-sRNA ASO regulatory activity in a cellular environment. The output from the pipeline was an anti-sRNA ASO that targets sRNAs to inhibit sRNA-mRNA interactions and modulate gene expression. This anti-sRNA ASO has potential for reprogramming gene expression for synthetic biology and/or antibacterial purposes. We anticipate that this pipeline will find widespread use in fields targeting RNA-based interactions as modulators of gene expression.
Collapse
Affiliation(s)
- Charlotte A. Henderson
- School of Biological Sciences and Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DY, United Kingdom
| | - Helen A. Vincent
- School of Biological Sciences and Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DY, United Kingdom
| | - Anastasia J. Callaghan
- School of Biological Sciences and Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DY, United Kingdom
| |
Collapse
|
33
|
Sousa SA, Feliciano JR, Pita T, Soeiro CF, Mendes BL, Alves LG, Leitão JH. Bacterial Nosocomial Infections: Multidrug Resistance as a Trigger for the Development of Novel Antimicrobials. Antibiotics (Basel) 2021; 10:antibiotics10080942. [PMID: 34438992 PMCID: PMC8389044 DOI: 10.3390/antibiotics10080942] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 11/17/2022] Open
Abstract
Nosocomial bacterial infections are associated with high morbidity and mortality, posing a huge burden to healthcare systems worldwide. The ongoing COVID-19 pandemic, with the raised hospitalization of patients and the increased use of antimicrobial agents, boosted the emergence of difficult-to-treat multidrug-resistant (MDR) bacteria in hospital settings. Therefore, current available antibiotic treatments often have limited or no efficacy against nosocomial bacterial infections, and novel therapeutic approaches need to be considered. In this review, we analyze current antibacterial alternatives under investigation, focusing on metal-based complexes, antimicrobial peptides, and antisense antimicrobial therapeutics. The association of new compounds with older, commercially available antibiotics and the repurposing of existing drugs are also revised in this work.
Collapse
Affiliation(s)
- Sílvia A. Sousa
- Department of Bioengineering, IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (J.R.F.); (T.P.); (C.F.S.); (B.L.M.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Correspondence: (S.A.S.); (J.H.L.); Tel.: +351-218417688 (J.H.L.)
| | - Joana R. Feliciano
- Department of Bioengineering, IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (J.R.F.); (T.P.); (C.F.S.); (B.L.M.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Tiago Pita
- Department of Bioengineering, IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (J.R.F.); (T.P.); (C.F.S.); (B.L.M.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Catarina F. Soeiro
- Department of Bioengineering, IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (J.R.F.); (T.P.); (C.F.S.); (B.L.M.)
| | - Beatriz L. Mendes
- Department of Bioengineering, IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (J.R.F.); (T.P.); (C.F.S.); (B.L.M.)
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Luis G. Alves
- Centro de Química Estrutural, Associação do Instituto Superior Técnico para a Investigação e Desenvolvimento, 1049-003 Lisboa, Portugal;
| | - Jorge H. Leitão
- Department of Bioengineering, IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (J.R.F.); (T.P.); (C.F.S.); (B.L.M.)
- Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Correspondence: (S.A.S.); (J.H.L.); Tel.: +351-218417688 (J.H.L.)
| |
Collapse
|
34
|
Popella L, Jung J, Popova K, Ðurica-Mitić S, Barquist L, Vogel J. Global RNA profiles show target selectivity and physiological effects of peptide-delivered antisense antibiotics. Nucleic Acids Res 2021; 49:4705-4724. [PMID: 33849070 PMCID: PMC8096218 DOI: 10.1093/nar/gkab242] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
Antisense peptide nucleic acids (PNAs) inhibiting mRNAs of essential genes provide a straight-forward way to repurpose our knowledge of bacterial regulatory RNAs for development of programmable species-specific antibiotics. While there is ample proof of PNA efficacy, their target selectivity and impact on bacterial physiology are poorly understood. Moreover, while antibacterial PNAs are typically designed to block mRNA translation, effects on target mRNA levels are not well-investigated. Here, we pioneer the use of global RNA-seq analysis to decipher PNA activity in a transcriptome-wide manner. We find that PNA-based antisense oligomer conjugates robustly decrease mRNA levels of the widely-used target gene, acpP, in Salmonella enterica, with limited off-target effects. Systematic analysis of several different PNA-carrier peptides attached not only shows different bactericidal efficiency, but also activation of stress pathways. In particular, KFF-, RXR- and Tat-PNA conjugates especially induce the PhoP/Q response, whereas the latter two additionally trigger several distinct pathways. We show that constitutive activation of the PhoP/Q response can lead to Tat-PNA resistance, illustrating the utility of RNA-seq for understanding PNA antibacterial activity. In sum, our study establishes an experimental framework for the design and assessment of PNA antimicrobials in the long-term quest to use these for precision editing of microbiota.
Collapse
Affiliation(s)
- Linda Popella
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080 Würzburg, Germany
| | - Jakob Jung
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080 Würzburg, Germany
| | - Kristina Popova
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080 Würzburg, Germany
| | - Svetlana Ðurica-Mitić
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080 Würzburg, Germany
| | - Lars Barquist
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), D-97080 Würzburg, Germany.,Faculty of Medicine, University of Würzburg, D-97080 Würzburg, Germany
| | - Jörg Vogel
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, D-97080 Würzburg, Germany.,Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), D-97080 Würzburg, Germany.,Faculty of Medicine, University of Würzburg, D-97080 Würzburg, Germany
| |
Collapse
|
35
|
Liang X, Liu M, Komiyama M. Recognition of Target Site in Various Forms of DNA and RNA by Peptide Nucleic Acid (PNA): From Fundamentals to Practical Applications. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2021. [DOI: 10.1246/bcsj.20210086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Xingguo Liang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, P. R. China
| | - Mengqin Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
| | - Makoto Komiyama
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P. R. China
| |
Collapse
|
36
|
Barbosa A, Araújo D, Henriques M, Silva S. The combined application of the anti-RAS1 and anti-RIM101 2'-OMethylRNA oligomers enhances Candida albicans filamentation control. Med Mycol 2021; 59:1024-1031. [PMID: 34097057 DOI: 10.1093/mmy/myab033] [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: 02/19/2021] [Revised: 05/12/2021] [Accepted: 06/02/2021] [Indexed: 01/30/2023] Open
Abstract
Whereas antisense oligomers (ASOs) have been successfully utilized to control gene expression, they have been little exploited to control Candida albicans virulence's determinants. Filamentation is an important virulence factor of C. albicans, and RAS1 and RIM101 genes are involved in its regulation. Thus, the main goal of this work was to project ASOs, based on 2'-OMethyl chemical modification, to target RAS1 and RIM101 mRNA and to validate its application either alone or in combination, to reduce Candida filamentation in different human body fluids.It was verified that both, anti-RAS1 2'OMe and anti-RIM101 2'OMe oligomers, were able to reduce the levels of RAS1 and RIM101 genes' expression and to significantly reduce C. albicans filamentation. Furthermore, the combined application of anti-RAS1 2'OMe oligomer and anti-RIM101 2'OMe oligomer enhances the control of C. albicans filamentation in artificial saliva and urine.Our work confirms that ASOs are useful tools for research and therapeutic development on the control of candidiasis. LAY ABSTRACT This work aimed to project antisense oligomers to control Candida albicans filamentation. The results revealed that the projected oligomers, anti-RAS1 2'OMe and anti-RIM101 2'OMe, were able to control RAS1 and RIM101 gene expression and to significantly reduce C. albicans filamentation.
Collapse
Affiliation(s)
- Ana Barbosa
- LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Daniela Araújo
- LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Mariana Henriques
- LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Sónia Silva
- LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
37
|
Goddard LR, Mardle CE, Gneid H, Ball CG, Gowers DM, Atkins HS, Butt LE, Watts JK, Vincent HA, Callaghan AJ. An Investigation into the Potential of Targeting Escherichia coli rne mRNA with Locked Nucleic Acid (LNA) Gapmers as an Antibacterial Strategy. Molecules 2021; 26:molecules26113414. [PMID: 34200016 PMCID: PMC8200214 DOI: 10.3390/molecules26113414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
The increase in antibacterial resistance is a serious challenge for both the health and defence sectors and there is a need for both novel antibacterial targets and antibacterial strategies. RNA degradation and ribonucleases, such as the essential endoribonuclease RNase E, encoded by the rne gene, are emerging as potential antibacterial targets while antisense oligonucleotides may provide alternative antibacterial strategies. As rne mRNA has not been previously targeted using an antisense approach, we decided to explore using antisense oligonucleotides to target the translation initiation region of the Escherichia coli rne mRNA. Antisense oligonucleotides were rationally designed and were synthesised as locked nucleic acid (LNA) gapmers to enable inhibition of rne mRNA translation through two mechanisms. Either LNA gapmer binding could sterically block translation and/or LNA gapmer binding could facilitate RNase H-mediated cleavage of the rne mRNA. This may prove to be an advantage over the majority of previous antibacterial antisense oligonucleotide approaches which used oligonucleotide chemistries that restrict the mode-of-action of the antisense oligonucleotide to steric blocking of translation. Using an electrophoretic mobility shift assay, we demonstrate that the LNA gapmers bind to the translation initiation region of E. coli rne mRNA. We then use a cell-free transcription translation reporter assay to show that this binding is capable of inhibiting translation. Finally, in an in vitro RNase H cleavage assay, the LNA gapmers facilitate RNase H-mediated mRNA cleavage. Although the challenges of antisense oligonucleotide delivery remain to be addressed, overall, this work lays the foundations for the development of a novel antibacterial strategy targeting rne mRNA with antisense oligonucleotides.
Collapse
Affiliation(s)
- Layla R. Goddard
- School of Biological Sciences and Institute of Biological & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DY, UK; (L.R.G.); (C.E.M.); (C.G.B.); (D.M.G.); (L.E.B.)
- Centre for Enzyme Innovation, University of Portsmouth, Portsmouth PO1 2DY, UK
| | - Charlotte E. Mardle
- School of Biological Sciences and Institute of Biological & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DY, UK; (L.R.G.); (C.E.M.); (C.G.B.); (D.M.G.); (L.E.B.)
| | - Hassan Gneid
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01609, USA; (H.G.); (J.K.W.)
- Department of Chemistry, University of Southampton, Southampton SO17 1BJ, UK
| | - Ciara G. Ball
- School of Biological Sciences and Institute of Biological & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DY, UK; (L.R.G.); (C.E.M.); (C.G.B.); (D.M.G.); (L.E.B.)
- Centre for Enzyme Innovation, University of Portsmouth, Portsmouth PO1 2DY, UK
| | - Darren M. Gowers
- School of Biological Sciences and Institute of Biological & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DY, UK; (L.R.G.); (C.E.M.); (C.G.B.); (D.M.G.); (L.E.B.)
| | - Helen S. Atkins
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK;
- College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Louise E. Butt
- School of Biological Sciences and Institute of Biological & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DY, UK; (L.R.G.); (C.E.M.); (C.G.B.); (D.M.G.); (L.E.B.)
| | - Jonathan K. Watts
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01609, USA; (H.G.); (J.K.W.)
| | - Helen A. Vincent
- School of Biological Sciences and Institute of Biological & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DY, UK; (L.R.G.); (C.E.M.); (C.G.B.); (D.M.G.); (L.E.B.)
- Centre for Enzyme Innovation, University of Portsmouth, Portsmouth PO1 2DY, UK
- Correspondence: (H.A.V.); (A.J.C.)
| | - Anastasia J. Callaghan
- School of Biological Sciences and Institute of Biological & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DY, UK; (L.R.G.); (C.E.M.); (C.G.B.); (D.M.G.); (L.E.B.)
- Centre for Enzyme Innovation, University of Portsmouth, Portsmouth PO1 2DY, UK
- Correspondence: (H.A.V.); (A.J.C.)
| |
Collapse
|
38
|
Farahani NN, Kalani BS, Monavari SH, Mirkalantari S, Montazer F, Sholeh M, Javanmard Z, Irajian G. Therapeutic effects, immunogenicity and cytotoxicity of a cell penetrating peptide-peptide nucleic acid conjugate against cagA of Helicobacter pylori in cell culture and animal model. IRANIAN JOURNAL OF MICROBIOLOGY 2021; 13:360-371. [PMID: 34540175 PMCID: PMC8416595 DOI: 10.18502/ijm.v13i3.6399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVES Helicobacter pylori causes several gastrointestinal diseases, including asymptomatic gastritis, chronic peptic ulcer, duodenal ulcer, lymphoma of the mucosa-associated lymphoid tissue (MALT), and gastric adenocarcinoma. In recent years, failure to eradicate H. pylori infections has become an alarming problem for physicians. It is now clear that the current treatment strategies may become ineffective, necessitating the development of innovative antimicrobial compounds as alternative treatments. MATERIALS AND METHODS In this experimental study, a cell-penetrating peptide-conjugated peptide nucleic acid (CPP-PNA) was used to target the cagA expression. cagA expression was evaluated using RT-qPCR assay after treatment by the CPPPNA in cell culture and animal model. Additionally, immunogenicity and toxicity of the CPP-PNA were assessed in both cell culture and animal models. RESULTS Our analysis showed that cagA mRNA levels reduced in H. pylori-infected HT29 cells after treatment with CPPPNA in a dose-dependent manner. Also, cagA expression in bacterial RNA extracted from stomach tissue of mice treated with PNA was reduced compared to that of untreated mice. The expression of inflammatory cytokines also decreased in cells and tissue of H. pylori-infected mice after PNA treatment. The tested CPP-PNA showed no significant adverse effects on cell proliferation of cultured cells and no detectable toxicity and immunogenicity were observed in mice. CONCLUSION These results suggest the effectiveness of CPP-PNA in targeting CagA for various research and therapeutic purposes, offering a potential antisense therapy against H. pylori infections.
Collapse
Affiliation(s)
- Narges Nodeh Farahani
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behrooz Sadeghi Kalani
- Department of Medical Microbiology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Shiva Mirkalantari
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Montazer
- Firoozabadi Clinical Research and Development Unit (FACRDU), Iran university of Medical Sciences (IUMS), Tehran, Iran
| | - Mohammad Sholeh
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Javanmard
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Irajian
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
da Silva KE, Ribeiro SM, Rossato L, Dos Santos CP, Preza SE, Cardoso MH, Franco OL, Migliolo L, Simionatto S. Antisense peptide nucleic acid inhibits the growth of KPC-producing Klebsiella pneumoniae strain. Res Microbiol 2021; 172:103837. [PMID: 34029675 DOI: 10.1016/j.resmic.2021.103837] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 11/17/2022]
Abstract
Klebsiella pneumoniae causes common and severe hospital- and community-acquired infections with a high incidence of multidrug resistance (MDR) and mortality. In this study, we investigated the ability of the antisense peptide nucleic acids (PNA) conjugated to the (KFF)3K cell-penetrating peptide (CPP) to target the gyrA KPC-producing K. pneumoniae and inhibit bacterial growth in vitro. The inhibitory effect on gyrA gene was evaluated by measuring 16s gene amplification in KPC-producing K. pneumoniae treated with the antisense PNA conjugate. The hemolytic property of the antisense PNA conjugate was accessed toward mice red blood cells. Finally, molecular modeling and dynamics simulations analyses in aqueous solutions were performed to predict the PNA conformation alone in contact with DNA (gyrA gene sequence). PNA was capable of inhibiting bacterial growth at 50 μM, also reducing 16S gene amplification in 96.7%. Besides, PNA presented low hemolytic activity (21.1% hemolysis) at this same concentration. Bioinformatics analysis demonstrated that the structure of the PNA is stable in water without major changes in its secondary structure. The ability of PNA and its conjugated CPP ((KFF)3K) to inhibit bacterial growth demonstrates the potential of this new class of antibacterial agents, encouraging further in vivo studies to confirm its therapeutic efficacy.
Collapse
Affiliation(s)
- Kesia Esther da Silva
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil.
| | - Suzana Meira Ribeiro
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil.
| | - Luana Rossato
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil.
| | - Caroline Paes Dos Santos
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil.
| | - Sergio Espindola Preza
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117900, Brazil.
| | - Marlon Henrique Cardoso
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117900, Brazil.
| | - Octávio Luiz Franco
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117900, Brazil; Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, 70790160, Brazil.
| | - Ludovico Migliolo
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117900, Brazil.
| | - Simone Simionatto
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil.
| |
Collapse
|
40
|
Araújo D, Braz J, Dencheva NV, Carvalho I, Henriques M, Denchev ZZ, Malfois M, Silva S. Polyamide Microsized Particulate Polyplex Carriers for the 2'- OMethylRNA EFG1 Antisense Oligonucleotide. ACS APPLIED BIO MATERIALS 2021; 4:4607-4617. [PMID: 35006798 DOI: 10.1021/acsabm.1c00334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Anti-EFG1 2'-OMethylRNA is an antisense oligonucleotide (ASO) that has the ability to recognize and block the EFG1 gene and to control Candida albicans filamentation. However, it is important to protect the anti-EFG1 2'-OMethylRNA ASO from the environmental human body conditions and to ensure that they will be delivered to their site of action, and polyplex microparticles (MPs) represent a class of vehicles to ASO cargo with these functionalities. Thus, the goal of this work was to develop polyplexes based on porous poly(γ-butyrolactam) (PA4) or poly(ε-caprolactam) (PA6) MPs for the anti-EFG1 2'-OMethylRNA ASO cargo and delivery. Two types of polyplexes were prepared with payloads of anti-EFG1 2'-OMethylRNA molecules, either entrapped or immobilized on prefabricated polyamide MPs. Our data confirm that PA4 and PA6 polyplex MPs can be feasible carriers for anti-EFG1 2'-OMethylRNA ASO molecules, using either the entrapment or immobilization strategies, whereby the released ASO maintains its activity against C. albicans cells.
Collapse
Affiliation(s)
- Daniela Araújo
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Joana Braz
- IPC-Institute for Polymers and Composites, Department of Polymers Engineering, University of Minho, 4800-056 Guimarães, Portugal
| | - Nadya V Dencheva
- IPC-Institute for Polymers and Composites, Department of Polymers Engineering, University of Minho, 4800-056 Guimarães, Portugal
| | - Isabel Carvalho
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Mariana Henriques
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Zlatan Z Denchev
- IPC-Institute for Polymers and Composites, Department of Polymers Engineering, University of Minho, 4800-056 Guimarães, Portugal
| | - Marc Malfois
- ALBA Synchrotron Facility, Cerdanyola del Valés, Barcelona 0890, Spain
| | - Sónia Silva
- LIBRO-Laboratório de Investigação em Biofilmes Rosário Oliveira, CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
41
|
Mastering the Gram-negative bacterial barrier - Chemical approaches to increase bacterial bioavailability of antibiotics. Adv Drug Deliv Rev 2021; 172:339-360. [PMID: 33705882 DOI: 10.1016/j.addr.2021.02.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/08/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
To win the battle against resistant, pathogenic bacteria, novel classes of anti-infectives and targets are urgently needed. Bacterial uptake, distribution, metabolic and efflux pathways of antibiotics in Gram-negative bacteria determine what we here refer to as bacterial bioavailability. Understanding these mechanisms from a chemical perspective is essential for anti-infective activity and hence, drug discovery as well as drug delivery. A systematic and critical discussion of in bacterio, in vitro and in silico assays reveals that a sufficiently accurate holistic approach is still missing. We expect new findings based on Gram-negative bacterial bioavailability to guide future anti-infective research.
Collapse
|
42
|
Targeting of the Essential acpP, ftsZ, and rne Genes in Carbapenem-Resistant Acinetobacter baumannii by Antisense PNA Precision Antibacterials. Biomedicines 2021; 9:biomedicines9040429. [PMID: 33921011 PMCID: PMC8071358 DOI: 10.3390/biomedicines9040429] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Infections by carbapenem-resistant A. baumannii (CRAB), a widespread nosocomial pathogen, are becoming increasingly difficult to prevent and treat. Therefore, there is an urgent need for discovery of novel antibiotics against CRAB. Programmable, precision antisense antibiotics, e.g., based on the nucleic acid mimic PNA (peptide nucleic acid) have shown promise in this respect in the form of PNA-BPP (bacteria penetrating peptide) conjugates targeting essential bacterial genes. In the present study, we designed and synthesized a series of PNA-BPPs targeting the translation initiation region of the ftsZ, acpP, or rne gene of CRAB strains. The antimicrobial activity of the compounds and effects on gene expression level was compared to that of analogous mismatch PNA controls. Three antisense conjugates (KFF)3K-eg1-(acpP)PNA (5639), (KFF)3K-eg1-(ftsZ)PNA (5612), and (KFF)3-K-eg1-(rne)PNA (5656) exhibited complete growth inhibition against several CRAB strains at 1-2, 2-8, and 2 µM, respectively, and the compounds were bactericidal at 1-2× MIC. The bactericidal effect was correlated to reduction of target gene mRNA level using RT-qPCR, and the compounds showed no bacterial membrane disruption activity at 1-2× MIC. PNA5612 was tested against a series of 12 CRAB isolates and all were sensitive at 2-8 µM. In addition, the conjugates exhibited no cellular toxicity in the HepG2 cell line (up to 20 μM) and did not shown significant antibacterial activity against other Gram negatives (E. coli, P. aeruginosa). These results provide a starting point for discovery of antisense precision designer antibiotics for specific treatment of CRAB infections.
Collapse
|
43
|
Silencing Antibiotic Resistance with Antisense Oligonucleotides. Biomedicines 2021; 9:biomedicines9040416. [PMID: 33921367 PMCID: PMC8068983 DOI: 10.3390/biomedicines9040416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 02/06/2023] Open
Abstract
Antisense technologies consist of the utilization of oligonucleotides or oligonucleotide analogs to interfere with undesirable biological processes, commonly through inhibition of expression of selected genes. This field holds a lot of promise for the treatment of a very diverse group of diseases including viral and bacterial infections, genetic disorders, and cancer. To date, drugs approved for utilization in clinics or in clinical trials target diseases other than bacterial infections. Although several groups and companies are working on different strategies, the application of antisense technologies to prokaryotes still lags with respect to those that target other human diseases. In those cases where the focus is on bacterial pathogens, a subset of the research is dedicated to produce antisense compounds that silence or reduce expression of antibiotic resistance genes. Therefore, these compounds will be adjuvants administered with the antibiotic to which they reduce resistance levels. A varied group of oligonucleotide analogs like phosphorothioate or phosphorodiamidate morpholino residues, as well as peptide nucleic acids, locked nucleic acids and bridge nucleic acids, the latter two in gapmer configuration, have been utilized to reduce resistance levels. The major mechanisms of inhibition include eliciting cleavage of the target mRNA by the host’s RNase H or RNase P, and steric hindrance. The different approaches targeting resistance to β-lactams include carbapenems, aminoglycosides, chloramphenicol, macrolides, and fluoroquinolones. The purpose of this short review is to summarize the attempts to develop antisense compounds that inhibit expression of resistance to antibiotics.
Collapse
|
44
|
Chen W, Dong B, Liu W, Liu Z. Recent Advances in Peptide Nucleic Acids as Antibacterial Agents. Curr Med Chem 2021; 28:1104-1125. [PMID: 32484766 DOI: 10.2174/0929867327666200602132504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/06/2020] [Accepted: 05/13/2020] [Indexed: 11/22/2022]
Abstract
The emergence of antibiotic-resistant bacteria and the slow progress in searching for new antimicrobial agents makes it hard to treat bacterial infections and cause problems for the healthcare system worldwide, including high costs, prolonged hospitalizations, and increased mortality. Therefore, the discovery of effective antibacterial agents is of great importance. One attractive alternative is antisense peptide nucleic acid (PNA), which inhibits or eliminates gene expression by binding to the complementary messenger RNA (mRNA) sequence of essential genes or the accessible and functionally important regions of the ribosomal RNA (rRNA). Following 30 years of development, PNAs have played an extremely important role in the treatment of Gram-positive, Gram-negative, and acidfast bacteria due to their desirable stability of hybrid complex with target RNA, the strong affinity for target mRNA/rRNA, and the stability against nucleases. PNA-based antisense antibiotics can strongly inhibit the growth of pathogenic and antibiotic-resistant bacteria in a sequence-specific and dose-dependent manner at micromolar concentrations. However, several fundamental challenges, such as intracellular delivery, solubility, physiological stability, and clearance still need to be addressed before PNAs become broadly applicable in clinical settings. In this review, we summarize the recent advances in PNAs as antibacterial agents and the challenges that need to be overcome in the future.
Collapse
Affiliation(s)
- Wei Chen
- Hunan Key Laboratory for Super Microstructure and Ultrafast Process, School of Physics and Electronics Central South University, Changsha 410083, China
| | - Bo Dong
- Hunan Key Laboratory for Super Microstructure and Ultrafast Process, School of Physics and Electronics Central South University, Changsha 410083, China
| | - Wenen Liu
- Department of Clinical Laboratory, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Zhengchun Liu
- Hunan Key Laboratory for Super Microstructure and Ultrafast Process, School of Physics and Electronics Central South University, Changsha 410083, China
| |
Collapse
|
45
|
Eller KA, Aunins TR, Courtney CM, Campos JK, Otoupal PB, Erickson KE, Madinger NE, Chatterjee A. Facile accelerated specific therapeutic (FAST) platform develops antisense therapies to counter multidrug-resistant bacteria. Commun Biol 2021; 4:331. [PMID: 33712689 PMCID: PMC7955031 DOI: 10.1038/s42003-021-01856-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/11/2021] [Indexed: 02/08/2023] Open
Abstract
Multidrug-resistant (MDR) bacteria pose a grave concern to global health, which is perpetuated by a lack of new treatments and countermeasure platforms to combat outbreaks or antibiotic resistance. To address this, we have developed a Facile Accelerated Specific Therapeutic (FAST) platform that can develop effective peptide nucleic acid (PNA) therapies against MDR bacteria within a week. Our FAST platform uses a bioinformatics toolbox to design sequence-specific PNAs targeting non-traditional pathways/genes of bacteria, then performs in-situ synthesis, validation, and efficacy testing of selected PNAs. As a proof of concept, these PNAs were tested against five MDR clinical isolates: carbapenem-resistant Escherichia coli, extended-spectrum beta-lactamase Klebsiella pneumoniae, New Delhi Metallo-beta-lactamase-1 carrying Klebsiella pneumoniae, and MDR Salmonella enterica. PNAs showed significant growth inhibition for 82% of treatments, with nearly 18% of treatments leading to greater than 97% decrease. Further, these PNAs are capable of potentiating antibiotic activity in the clinical isolates despite presence of cognate resistance genes. Finally, the FAST platform offers a novel delivery approach to overcome limited transport of PNAs into mammalian cells by repurposing the bacterial Type III secretion system in conjunction with a kill switch that is effective at eliminating 99.6% of an intracellular Salmonella infection in human epithelial cells.
Collapse
Affiliation(s)
- Kristen A Eller
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Thomas R Aunins
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Colleen M Courtney
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- Sachi Bioworks, Inc, Boulder, CO, 80301, USA
| | - Jocelyn K Campos
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Peter B Otoupal
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Keesha E Erickson
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Nancy E Madinger
- Division of Infectious Diseases, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Anushree Chatterjee
- Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA.
- Sachi Bioworks, Inc, Boulder, CO, 80301, USA.
- Biomedical Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA.
- Antimicrobial Regeneration Consortium, Boulder, CO, 80301, USA.
| |
Collapse
|
46
|
Streicher LM. Exploring the future of infectious disease treatment in a post-antibiotic era: A comparative review of alternative therapeutics. J Glob Antimicrob Resist 2021; 24:285-295. [PMID: 33484895 DOI: 10.1016/j.jgar.2020.12.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/19/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022] Open
Abstract
Antibiotic resistance is projected to be one of the greatest healthcare challenges of the 21st century. As the efficacy of these critical drugs wanes and the discovery of new antibiotics stagnates, exploration of alternative therapies could offer a much needed solution. Although numerous alternative therapies are currently under investigation, three in particular appear poised for long-term success, namely antimicrobial oligonucleotides, monoclonal antibodies and phage therapy. Antimicrobial oligonucleotides could conceivably offer the greatest spectrum of activity while having the lowest chance of unrecoverable resistance. Bacteriophages, while most susceptible to resistance, are inexhaustible, inexpensive and exceptionally adept at eliminating biofilm-associated infections. And although monoclonal antibodies may have limited access to such recalcitrant bacteria, these agents are uniquely able to neutralise exotoxins and other diffusible virulence factors. This comparative review seeks to illuminate these promising therapies and to encourage the scientific and financial support necessary to usher in the next generation of infectious disease treatment.
Collapse
|
47
|
Abstract
Vitamin B12 (cobalamin, Cbl) is an essential nutrient for all mammals and some bacteria. From a chemical point of view, it is a highly functionalized molecule, which enables conjugation at multiple positions and attachment of various cargoes. Both mammalian and bacterial cells have developed a specific transport pathway for the uptake of vitamin B12, and as a consequence, cobalamin is an attractive candidate for the delivery of biologically relevant molecules into cells. Indeed, hybrid molecules containing vitamin B12 in their structure have found various applications in medicinal chemistry, diagnostics, and biological sciences.Herein, we describe synthetic strategies toward the synthesis of vitamin B12 conjugates with peptide nucleic acid (PNA ) oligomers. Such short-modified oligonucleotides targeted at bacterial DNA or RNA can act as antibacterial agents if efficiently taken up by bacterial cells. The uptake of such oligonucleotides is hindered by the bacterial cell envelope, but vitamin B12 was found to efficiently deliver antisense PNA into Escherichia coli and Salmonella Typhimurium cells. This paves the way to the use of vitamin B12-PNA conjugates in antibacterial and diagnostic applications.Vitamin B12-PNA conjugates can be prepared via copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) that gives access to covalently linked hybrids or via connecting both building blocks by reduction-sensitive disulfide bridge. Both approaches require prior modification of vitamin B12 by incorporation of the azide moiety or via transformation of the native functional group into a moiety reactive toward thiols. Conjugation of vitamin B12 with PNA-tagged substrates efficiently furnishes designed conjugates.
Collapse
Affiliation(s)
| | | | - Joanna Trylska
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Dorota Gryko
- Institute of Organic Chemistry, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
48
|
Vogel J. An RNA biology perspective on species-specific programmable RNA antibiotics. Mol Microbiol 2020; 113:550-559. [PMID: 32185839 DOI: 10.1111/mmi.14476] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
Our body is colonized by a vast array of bacteria the sum of which forms our microbiota. The gut alone harbors >1,000 bacterial species. An understanding of their individual or synergistic contributions to human health and disease demands means to interfere with their functions on the species level. Most of the currently available antibiotics are broad-spectrum, thus too unspecific for a selective depletion of a single species of interest from the microbiota. Programmable RNA antibiotics in the form of short antisense oligonucleotides (ASOs) promise to achieve precision manipulation of bacterial communities. These ASOs are coupled to small peptides that carry them inside the bacteria to silence mRNAs of essential genes, for example, to target antibiotic-resistant pathogens as an alternative to standard antibiotics. There is already proof-of-principle with diverse bacteria, but many open questions remain with respect to true species specificity, potential off-targeting, choice of peptides for delivery, bacterial resistance mechanisms and the host response. While there is unlikely a one-fits-all solution for all microbiome species, I will discuss how recent progress in bacterial RNA biology may help to accelerate the development of programmable RNA antibiotics for microbiome editing and other applications.
Collapse
Affiliation(s)
- Jörg Vogel
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), Würzburg, Germany.,RNA Biology Group, Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Davies JE, Behroozian S. An ancient solution to a modern problem. Mol Microbiol 2020; 113:546-549. [PMID: 32185831 DOI: 10.1111/mmi.14481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 11/30/2022]
Abstract
The dearth of new antibiotics and escalating emergence of multidrug resistant bacteria have created a global healthcare crisis and highlight the drastic need for novel antimicrobial agents. Complementary and alternative strategies including the investigation of ancient medicinals could address this problem. Natural clay minerals with a long history of medicinal and biomedical applications have become an interest due to their broad-spectrum antimicrobial activity. Such untapped natural sources may provide new therapeutic agents in the battle against infectious diseases in the post-antibiotic era.
Collapse
Affiliation(s)
- Julian E Davies
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Shekooh Behroozian
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
50
|
Piattelli E, Peltier J, Soutourina O. Interplay between Regulatory RNAs and Signal Transduction Systems during Bacterial Infection. Genes (Basel) 2020; 11:E1209. [PMID: 33081172 PMCID: PMC7602753 DOI: 10.3390/genes11101209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
The ability of pathogenic bacteria to stably infect the host depends on their capacity to respond and adapt to the host environment and on the efficiency of their defensive mechanisms. Bacterial envelope provides a physical barrier protecting against environmental threats. It also constitutes an important sensory interface where numerous sensing systems are located. Signal transduction systems include Two-Component Systems (TCSs) and alternative sigma factors. These systems are able to sense and respond to the ever-changing environment inside the host, altering the bacterial transcriptome to mitigate the impact of the stress. The regulatory networks associated with signal transduction systems comprise small regulatory RNAs (sRNAs) that can be directly involved in the expression of virulence factors. The aim of this review is to describe the importance of TCS- and alternative sigma factor-associated sRNAs in human pathogens during infection. The currently available genome-wide approaches for studies of TCS-regulated sRNAs will be discussed. The differences in the signal transduction mediated by TCSs between bacteria and higher eukaryotes and the specificity of regulatory RNAs for their targets make them appealing targets for discovery of new strategies to fight against multi-resistant bacteria.
Collapse
Affiliation(s)
- Emma Piattelli
- Institute for Integrative Biology of the Cell (I2BC), CNRS, CEA, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (E.P.); (J.P.)
| | - Johann Peltier
- Institute for Integrative Biology of the Cell (I2BC), CNRS, CEA, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (E.P.); (J.P.)
- Laboratoire Pathogenèses des Bactéries Anaérobies, Institut Pasteur, UMR CNRS 2001, Université de Paris, 75015 Paris, France
| | - Olga Soutourina
- Institute for Integrative Biology of the Cell (I2BC), CNRS, CEA, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (E.P.); (J.P.)
- Institut Universitaire de France, CEDEX 05, 75231 Paris, France
| |
Collapse
|