1
|
Pals MJ, Lindberg A, Velema WA. Chemical strategies for antisense antibiotics. Chem Soc Rev 2024. [PMID: 39436264 PMCID: PMC11495246 DOI: 10.1039/d4cs00238e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
2
|
Faghirabadi F, Abuei H, Malekzadeh MH, Mojiri A, Farhadi A. Intracellular delivery of antiviral shRNA using penetratin-based complexes effectively inhibits respiratory syncytial virus replication and host cell apoptosis. Virol J 2024; 21:235. [PMID: 39350281 PMCID: PMC11443668 DOI: 10.1186/s12985-024-02519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Cell-penetrating peptides (CPPs) are effective for delivering therapeutic molecules with minimal toxicity. This study focuses on the use of penetratin, a well-characterized CPP, to deliver a DNA vector encoding short hairpin RNA (shRNA) targeting the respiratory syncytial virus (RSV) F gene into infected cells. RSV is known to cause severe lower respiratory infections in infants and poses significant risks to immunocompromised individuals and the elderly. We evaluated the antiviral efficacy of the penetratin-shRNA complex by comparing its ability to inhibit RSV replication and induce apoptosis with ribavirin treatment. METHODS Penetratin-shRNA complexes were prepared at different ratios and analyzed using gel retardation assays, dynamic light scattering, and zeta potential measurements. The complexes were tested in HEp-2 and A549 cells for transfection efficiency, cytotoxicity, viral load, and apoptosis using plaque assays, real-time reverse transcription-polymerase chain reaction (RT-PCR), DNA fragmentation, propidium iodide staining, and caspase 3/7 activation assays. RESULTS The gel shift assay determined that a 20:1 CPP-to-shRNA ratio was optimal for effective complexation, resulting in particles with a size of 164 nm and a zeta potential of 8.7 mV. Transfection efficiency in HEp-2 cells was highest at this ratio, reaching up to 93%. The penetratin-shRNA complex effectively silenced the RSV F gene, reduced viral titers, and decreased DNA fragmentation and apoptosis in infected cells. CONCLUSION Penetratin effectively delivers shRNA targeting the RSV F gene, significantly reducing viral load and preventing apoptosis without toxicity. This approach surpasses Lipofectamine and shows potential for future therapeutic interventions, especially when combined with ribavirin, against RSV infection.
Collapse
Affiliation(s)
- Faezeh Faghirabadi
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Haniyeh Abuei
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Malekzadeh
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Anahita Mojiri
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, 77030, TX, USA
| | - Ali Farhadi
- Division of Medical Biotechnology, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Lopuszynski J, Wang J, Zahid M. Beyond Transduction: Anti-Inflammatory Effects of Cell Penetrating Peptides. Molecules 2024; 29:4088. [PMID: 39274936 PMCID: PMC11397606 DOI: 10.3390/molecules29174088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
One of the bottlenecks to bringing new therapies to the clinic has been a lack of vectors for delivering novel therapeutics in a targeted manner. Cell penetrating peptides (CPPs) have received a lot of attention and have been the subject of numerous developments since their identification nearly three decades ago. Known for their transduction abilities, they have generally been considered inert vectors. In this review, we present a schema for their classification, highlight what is known about their mechanism of transduction, and outline the existing literature as well as our own experience, vis a vis the intrinsic anti-inflammatory properties that certain CPPs exhibit. Given the inflammatory responses associated with viral vectors, CPPs represent a viable alternative to such vectors; furthermore, the anti-inflammatory properties of CPPs, mostly through inhibition of the NF-κB pathway, are encouraging. Much more work in relevant animal models, toxicity studies in large animal models, and ultimately human trials are needed before their potential is fully realized.
Collapse
Affiliation(s)
| | | | - Maliha Zahid
- Department of Cardiovascular Medicine, Guggenheim Gu 9-01B, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
4
|
Qian K, Yang P, Li Y, Meng R, Cheng Y, Zhou L, Wu J, Xu S, Bao X, Guo Q, Wang P, Xu M, Sheng D, Zhang Q. Rational fusion design inspired by cell-penetrating peptide: SS31/S-14 G Humanin hybrid peptide with amplified multimodal efficacy and bio-permeability for the treatment of Alzheimer's disease. Asian J Pharm Sci 2024; 19:100938. [PMID: 39253611 PMCID: PMC11382307 DOI: 10.1016/j.ajps.2024.100938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/26/2023] [Accepted: 05/16/2024] [Indexed: 09/11/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease induced by multiple interconnected mechanisms. Peptide drug candidates with multi-modal efficacy generated from fusion strategy are suitable for addressing multi-facet pathology. However, clinical translation of peptide drugs is greatly hampered by their low permeability into brain. Herein, a hybrid peptide HNSS is generated by merging two therapeutic peptides (SS31 and S-14 G Humanin (HNG)), using a different approach from the classical shuttle-therapeutic peptide conjugate design. HNSS demonstrated increased bio-permeability, with a 2-fold improvement in brain distribution over HNG, thanks to its structure mimicking the design of signal peptide-derived cell-penetrating peptides. HNSS efficiently alleviated mitochondrial dysfunction through the combined effects of mitochondrial targeting, ROS scavenging and p-STAT3 activation. Meanwhile, HNSS with increased Aβ affinity greatly inhibited Aβ oligomerization/fibrillation, and interrupted Aβ interaction with neuron/microglia by reducing neuronal mitochondrial Aβ deposition and promoting microglial phagocytosis of Aβ. In 3× Tg-AD transgenic mice, HNSS treatment efficiently inhibited brain neuron loss and improved the cognitive performance. This work validates the rational fusion design-based strategy for bio-permeability improvement and efficacy amplification, providing a paradigm for developing therapeutic peptide candidates against neurodegenerative disease.
Collapse
Affiliation(s)
- Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuting Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiaoyan Bao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qian Guo
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
5
|
Ji K, Yao Y, Gao Y, Huang S, Ma L, Pan Q, Wu J, Zhang W, Chen H, Zhang L. Evaluating the cytotoxicity mechanism of the cell-penetrating peptide TP10 on Jurkat cells. Biochimie 2024; 221:182-192. [PMID: 37922978 DOI: 10.1016/j.biochi.2023.11.001] [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: 06/09/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
TP10, a classic cell-penetrating peptide, shows a high degree of similarity to AMPs in structure. Although TP10 has been widely used in drug delivery, the mechanism underlying its cytotoxicity is yet to be elucidated. Herein, we explored the cell-killing mechanism of TP10 against human leukemia Jurkat cells. TP10 induced necrosis in Jurkat cells via rapid disruption of cell membranes, particularly at high concentrations. Although mitochondria in Jurkat cells were damaged by TP10, mitochondria-mediated apoptosis did not occur, possibly due to intracellular ATP depletion. Necroptosis in TP10-treated Jurkat cells became an alternative route of apoptosis. Our results demonstrate that necrosis and necroptosis rather than apoptosis are involved in the cell-killing mechanism of TP10, which contributes to the understanding of its toxicity.
Collapse
Affiliation(s)
- Kun Ji
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Yufan Yao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yuxuan Gao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Sujie Huang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Ling Ma
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qing Pan
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Jun Wu
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Wei Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, 730000, China.
| | - Hongmei Chen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Lei Zhang
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Shi S, Zhang J, Quan S, Yang Y, Yao L, Xiao J. A highly biocompatible and bioactive transdermal nano collagen for enhanced healing of UV-damaged skin. Int J Biol Macromol 2024; 272:132857. [PMID: 38834124 DOI: 10.1016/j.ijbiomac.2024.132857] [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: 01/29/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 06/06/2024]
Abstract
Skin damage caused by excessive UV radiation has gradually become one of the most prevalent skin diseases. Collagen has gradually found applications in the treatment of UV-damaged skin; however, their high molecular weight greatly limits their capacity to permeate the skin barrier and repair the damaged skin. Nano collagen has garnered growing attentions in the mimicking of collagen; while the investigation of its skin permeability and wound-healing capability remains vacancies. Herein, we have for the first time created a highly biocompatible and bioactive transdermal nano collagen demonstrating remarkable transdermal capacity and repair efficacy for UV-damaged skin. The transdermal nano collagen exhibited a stable triple-helix structure, effectively promoting the adhesion and proliferation of fibroblasts. Notably, the transdermal nano collagen displayed exceptional penetration capabilities, permeating fibroblast and healthy skin. Combo evaluations revealed that the transdermal nano collagen contributed to recovering the intensity and TEWL values of UV-damaged skin to normal level. Histological analysis further indicated that transdermal nano collagen significantly accelerated the repair of damaged skin by promoting the collagen regeneration and fibroblasts activation. This highly biocompatible and bioactive transdermal nano collagen provides a novel substituted strategy for the transdermal absorption of collagen, indicating great potential applications in cosmetics and dermatology.
Collapse
Affiliation(s)
- Shuangni Shi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou 730000, PR China
| | - Jingting Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou 730000, PR China
| | - Siqi Quan
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou 730000, PR China
| | - Yi Yang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou 730000, PR China
| | - Linyan Yao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; School of Life Science, Lanzhou University, Lanzhou 730000, PR China.
| | - Jianxi Xiao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China; Gansu Engineering Research Center of Medical Collagen, Lanzhou 730000, PR China.
| |
Collapse
|
7
|
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
|
8
|
Chatterjee S, Kon E, Sharma P, Peer D. Endosomal escape: A bottleneck for LNP-mediated therapeutics. Proc Natl Acad Sci U S A 2024; 121:e2307800120. [PMID: 38437552 PMCID: PMC10945858 DOI: 10.1073/pnas.2307800120] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Lipid nanoparticles (LNPs) have recently emerged as a powerful and versatile clinically approved platform for nucleic acid delivery, specifically for mRNA vaccines. A major bottleneck in the field is the release of mRNA-LNPs from the endosomal pathways into the cytosol of cells where they can execute their encoded functions. The data regarding the mechanism of these endosomal escape processes are limited and contradicting. Despite extensive research, there is no consensus regarding the compartment of escape, the cause of the inefficient escape and are currently lacking a robust method to detect the escape. Here, we review the currently known mechanisms of endosomal escape and the available methods to study this process. We critically discuss the limitations and challenges of these methods and the possibilities to overcome these challenges. We propose that the development of currently lacking robust, quantitative high-throughput techniques to study endosomal escape is timely and essential. A better understanding of this process will enable better RNA-LNP designs with improved efficiency to unlock new therapeutic modalities.
Collapse
Affiliation(s)
- Sushmita Chatterjee
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Edo Kon
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Preeti Sharma
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
9
|
Kawaguchi Y, Kawamura Y, Hirose H, Kiyokawa M, Hirate M, Hirata T, Higuchi Y, Futaki S. E3MPH16: An efficient endosomolytic peptide for intracellular protein delivery. J Control Release 2024; 367:877-891. [PMID: 38301930 DOI: 10.1016/j.jconrel.2024.01.067] [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: 11/12/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
To facilitate the introduction of proteins, such as antibodies, into cells, a variety of delivery peptides have been engineered. These peptides are typically highly cationic and somewhat hydrophobic, enabling cytosolic protein delivery at the cost of causing cell damage by rupturing membranes. This balance between delivery effectiveness and cytotoxicity presents obstacles for their real-world use. To tackle this problem, we designed a new endosome-disruptive cytosolic delivery peptide, E3MPH16, inspired by mastoparan X (MP). E3MPH16 was engineered to incorporate three Glu (E3) and 16 His (H16) residues at the N- and C-termini of MP, respectively. The negative charges of E3 substantially mitigate the cell-surface damage induced by MP. The H16 segment is known to enhance cell-surface adsorption and endocytic uptake of the associated molecules. With these modifications, E3MPH16 was successfully trapped within endosomes. The acidification of endosomes is expected to protonate the side chains of E3 and H16, enabling E3MPH16 to rupture endosomal membranes. As a result, nearly 100% of cells achieved cytosolic delivery of a model biomacromolecule, Alexa Fluor 488-labeled dextran (10 kDa), via endosomal escape by co-incubation with E3MPH16. The delivery process also suggested the involvement of macropinocytosis and caveolae-mediated endocytosis. With the assistance of E3MPH16, Cre recombinase and anti-Ras-IgG delivered into HEK293 cells and HT1080 cells enabled gene recombination and inhibited cell proliferation, respectively. The potential for in vivo application of this intracellular delivery method was further validated by topically injecting the green fluorescent protein fused with a nuclear localization signal (NLS-GFP) along with E3MPH16 into Colon-26 tumor xenografts in mice.
Collapse
Affiliation(s)
- Yoshimasa Kawaguchi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Yuki Kawamura
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Hisaaki Hirose
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Megumi Kiyokawa
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Momo Hirate
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tsuyoshi Hirata
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuriko Higuchi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
10
|
Srivastava V, Godara P, Jena SP, Naik B, Singh S, Prajapati VK, Prusty D. Peptide-ligand conjugate based immunotherapeutic approach for targeted dismissal of non-structural protein 1 of dengue virus: A novel therapeutic solution for mild and severe dengue infections. Int J Biol Macromol 2024; 260:129562. [PMID: 38246445 DOI: 10.1016/j.ijbiomac.2024.129562] [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: 09/20/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Dengue virus infection has significantly increased, with reported cases soaring from 505,430 in 2000 to 2,809,818 in 2022, emphasizing the need for effective treatments. Among the eleven structural and non-structural proteins of DENV, Non-structural protein 1 (NS1) has emerged as a promising target due to its diverse role in modulating the immune response, inducing vascular leakage, and facilitating viral replication and assembly. Monoclonal antibodies are the sole therapeutics to target NS1, but concerns about their cross-reactivity persist. Given these concerns, our study focuses on designing a novel Peptide Ligand Conjugate (PLC) as a potential alternative immunotherapeutic agent against NS1. This PLC aims to mediate the immune elimination of soluble NS1 and NS1-presenting DENV-infected host cells by pre-existing vaccine-induced immunity. By employing the High Throughput Virtual Screening (HTVS) method, QikProp analysis, and Molecular Dynamics studies, we identified three hits from Asinex Biodesigned Ligands out of 220,177 compounds that show strong binding affinity towards the monoclonal binding site of NS1 protein. After a rigorous analysis of physicochemical characteristics, antigenicity, allergenicity, and toxicity using various servers, we selected two peptides: the minimum epitopic region of the Diphtheria and Tetanus toxins as the peptide components of the PLCs. A non-cleavable, non-reactive oxime linker connected the ligand with the peptide through oxime and amide bonds. DPT vaccine is widely used in dengue-endemic countries, and it has been reported that antibodies titer against MER of Diphtheria toxin and Tetanus toxins persist lifelong in DPT-vaccinated people. Therefore, once the rationally designed PLCs bind to NS1 through the ligands, the peptide will induce an immune response against NS1 by triggering pre-existing DPT antibodies and activating memory cells. This orchestrated immune response will destroy soluble NS1 and NS1-expressing DENV-infected cells, thereby reducing the illness of severe dengue hemorrhagic fever and the DENV infection, respectively. Given the increasing demand for new therapeutics for DENV treatment, further investigation into this novel immune-therapeutic strategy may offer a new avenue for treating mild and severe dengue infections.
Collapse
Affiliation(s)
- Varshita Srivastava
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Priya Godara
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Sudip Prasad Jena
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Biswajit Naik
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Satyendra Singh
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Dhaneswar Prusty
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India.
| |
Collapse
|
11
|
Shin HJ, Lee BK, Kang HA. Transdermal Properties of Cell-Penetrating Peptides: Applications and Skin Penetration Mechanisms. ACS APPLIED BIO MATERIALS 2024; 7:1-16. [PMID: 38079575 DOI: 10.1021/acsabm.3c00659] [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] [Indexed: 01/16/2024]
Abstract
Cell-penetrating peptides (CPPs) consist of 5-30 amino acids with intracellular transduction abilities and diverse physicochemical properties, origins, and sequences. Although recent developments in bioinformatics have facilitated the prediction of CPP candidates with the potential for transduction into cells, the mechanisms by which CPPs penetrate cells and various tissues have not yet been elucidated at the molecular interaction level. Recently, the skin-penetrating ability of CPPs has gained wide attention and emerged as a simple and effective strategy for the delivery of macromolecules into the skin. Studies on the skin structure have suggested that the penetration potential of CPPs is based on the molecular interactions and characteristics of the lipid lamellar structure between corneocytes in the stratum corneum. This review provides a brief overview of the general properties, transduction mechanisms, applications, and safety issues of CPPs, focusing on CPPs with transdermal properties, that are currently being used to develop therapeutics and cosmetics.
Collapse
Affiliation(s)
- Hee Je Shin
- ProCell R&D Center, ProCell Therapeutics, Inc., #1009 Ace-Twin Tower II, 273, Digital-ro, Guro-gu, Seoul 08381, Republic of Korea
- Department of Life Science, College of Natural Science, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Byung Kyu Lee
- ProCell R&D Center, ProCell Therapeutics, Inc., #1009 Ace-Twin Tower II, 273, Digital-ro, Guro-gu, Seoul 08381, Republic of Korea
| | - Hyun Ah Kang
- Department of Life Science, College of Natural Science, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
12
|
Sahagun DA, Lopuszynski JB, Feldman KS, Pogodzinski N, Zahid M. Toxicity Studies of Cardiac-Targeting Peptide Reveal a Robust Safety Profile. Pharmaceutics 2024; 16:73. [PMID: 38258084 PMCID: PMC10818749 DOI: 10.3390/pharmaceutics16010073] [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: 11/22/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
Targeted delivery of therapeutics specifically to cardiomyocytes would open up new frontiers for common conditions like heart failure. Our prior work using a phage display methodology identified a 12-amino-acid-long peptide that selectively targets cardiomyocytes after an intravenous injection in as little as 5 min and was hence termed a cardiac-targeting peptide (CTP: APHLSSQYSRT). CTP has been used to deliver imaging agents, small drug molecules, photosensitizing nanoparticles, exosomes, and even miRNA to cardiomyocytes. As a natural extension to the development of CTP as a clinically viable cardiac vector, we now present toxicity studies performed with the peptide. In vitro viability studies were performed in a human left ventricular myocyte cell line with 10 µM of Cyanine-5.5-labeled CTP (CTP-Cy5.5). In vitro ion channel profiles were completed for CTP followed by extensive studies in stably transfected cell lines for several GPCR-coupled receptors. Positive data for GPCR-coupled receptors were interrogated further with RT-qPCRs performed on mouse heart tissue. In vivo studies consisted of pre- and post-blood pressure monitoring acutely after a single CTP (10 mg/Kg) injection. Further in vivo toxicity studies consisted of injecting CTP (150 µg/Kg) in 60, 6-week-old, wild-type CD1, male/female mice (1:1), with cohorts of mice euthanized on days 0, 1, 2, 7, and 14 with inhalational CO2, followed by blood collection via cardiac puncture, complete blood count analysis, metabolic profiling, and finally, liver, renal, and thyroid studies. Lastly, mouse cardiac MRI was performed immediately before and after CTP (150 µg/Kg) injection to assess changes in cardiac size or function. Human left ventricular cardiomyocytes showed no decrease in viability after a 30 min incubation with CTP-Cy5.5. No significant activation or inhibition of any of seventy-eight protein channels was observed other than OPRM1 and COX2 at the highest tested concentration, neither of which were expressed in mouse heart tissue as assessed using RT-qPCR. CTP (10 mg/Kg) injections led to no change in blood pressure. Blood counts and chemistries showed no evidence of significant hematological, hepatic, or renal toxicities. Lastly, there was no difference in cardiac function, size, or mass acutely in response to CTP injections. Our studies with CTP showed no activation or inhibition of GPCR-associated receptors in vitro. We found no signals indicative of toxicity in vivo. Most importantly, cardiac functions remained unchanged acutely in response to CTP uptake. Further studies using good laboratory practices are needed with prolonged, chronic administration of CTP conjugated to a specific cargo of choice before human studies can be contemplated.
Collapse
Affiliation(s)
- Daniella A. Sahagun
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (D.A.S.); (J.B.L.)
| | - Jack B. Lopuszynski
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (D.A.S.); (J.B.L.)
| | - Kyle S. Feldman
- Clinical Virology Laboratory, Yale New Haven Hospital, New Haven, CT 06511, USA;
| | - Nicholas Pogodzinski
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Maliha Zahid
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (D.A.S.); (J.B.L.)
| |
Collapse
|
13
|
Lica JJ, Heldt M, Wieczór M, Chodnicki P, Ptaszyńska N, Maciejewska N, Łęgowska A, Brankiewicz W, Gucwa K, Stupak A, Pradhan B, Gitlin-Domagalska A, Dębowski D, Milewski S, Bieniaszewska M, Grabe GJ, Hellmann A, Rolka K. Dual-Activity Fluoroquinolone-Transportan 10 Conjugates Offer Alternative Leukemia Therapy during Hematopoietic Cell Transplantation. Mol Pharmacol 2023; 105:39-53. [PMID: 37977824 DOI: 10.1124/molpharm.123.000735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/01/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023] Open
Abstract
Hematopoietic cell transplantation (HCT) is often considered a last resort leukemia treatment, fraught with limited success due to microbial infections, a leading cause of mortality in leukemia patients. To address this critical issue, we explored a novel approach by synthesizing antileukemic agents containing antibacterial substances. This innovative strategy involves conjugating fluoroquinolone antibiotics, such as ciprofloxacin (CIP) or levofloxacin (LVX), with the cell-penetrating peptide transportan 10 (TP10). Here, we demonstrate that the resultant compounds display promising biologic activities in preclinical studies. These novel conjugates not only exhibit potent antimicrobial effects but are also selective against leukemia cells. The cytotoxic mechanism involves rapid disruption of cell membrane asymmetry leading to membrane damage. Importantly, these conjugates penetrated mammalian cells, accumulating within the nuclear membrane without significant effect on cellular architecture or mitochondrial function. Molecular simulations elucidated the aggregation tendencies of TP10 conjugates within lipid bilayers, resulting in membrane disruption and permeabilization. Moreover, mass spectrometry analysis confirmed efficient reduction of disulfide bonds within TP10 conjugates, facilitating release and activation of the fluoroquinolone derivatives. Intriguingly, these compounds inhibited human topoisomerases, setting them apart from traditional fluoroquinolones. Remarkably, TP10 conjugates generated lower intracellular levels of reactive oxygen species compared with CIP and LVX. The combination of antibacterial and antileukemic properties, coupled with selective cytostatic effects and minimal toxicity toward healthy cells, positions TP10 derivatives as promising candidates for innovative therapeutic approaches in the context of antileukemic HCT. This study highlights their potential in search of more effective leukemia treatments. SIGNIFICANCE STATEMENT: Fluoroquinolones are commonly used antibiotics, while transportan 10 (TP10) is a cell-penetrating peptide (CPP) with anticancer properties. In HCT, microbial infections are the primary cause of illness and death. Combining TP10 with fluoroquinolones enhanced their effects on different cell types. The dual pharmacological action of these conjugates offers a promising proof-of-concept solution for leukemic patients undergoing HCT. Strategically designed therapeutics, incorporating CPPs with antibacterial properties, have the potential to reduce microbial infections in the treatment of malignancies.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Mateusz Heldt
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Milosz Wieczór
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Pawel Chodnicki
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Natalia Ptaszyńska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Natalia Maciejewska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Anna Łęgowska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Wioletta Brankiewicz
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Katarzyna Gucwa
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Anna Stupak
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Bhaskar Pradhan
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Agata Gitlin-Domagalska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Dawid Dębowski
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Sławomir Milewski
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Maria Bieniaszewska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Grzegorz Jan Grabe
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Andrzej Hellmann
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Krzysztof Rolka
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| |
Collapse
|
14
|
Haque US, Yokota T. Enhancing Antisense Oligonucleotide-Based Therapeutic Delivery with DG9, a Versatile Cell-Penetrating Peptide. Cells 2023; 12:2395. [PMID: 37830609 PMCID: PMC10572411 DOI: 10.3390/cells12192395] [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: 08/26/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Antisense oligonucleotide-based (ASO) therapeutics have emerged as a promising strategy for the treatment of human disorders. Charge-neutral PMOs have promising biological and pharmacological properties for antisense applications. Despite their great potential, the efficient delivery of these therapeutic agents to target cells remains a major obstacle to their widespread use. Cellular uptake of naked PMO is poor. Cell-penetrating peptides (CPPs) appear as a possibility to increase the cellular uptake and intracellular delivery of oligonucleotide-based drugs. Among these, the DG9 peptide has been identified as a versatile CPP with remarkable potential for enhancing the delivery of ASO-based therapeutics due to its unique structural features. Notably, in the context of phosphorodiamidate morpholino oligomers (PMOs), DG9 has shown promise in enhancing delivery while maintaining a favorable toxicity profile. A few studies have highlighted the potential of DG9-conjugated PMOs in DMD (Duchenne Muscular Dystrophy) and SMA (Spinal Muscular Atrophy), displaying significant exon skipping/inclusion and functional improvements in animal models. The article provides an overview of a detailed understanding of the challenges that ASOs face prior to reaching their targets and continued advances in methods to improve their delivery to target sites and cellular uptake, focusing on DG9, which aims to harness ASOs' full potential in precision medicine.
Collapse
Affiliation(s)
- Umme Sabrina Haque
- Department of Neuroscience, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Science Research, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
15
|
Zeng Y, Shen M, Pattipeiluhu R, Zhou X, Zhang Y, Bakkum T, Sharp TH, Boyle AL, Kros A. Efficient mRNA delivery using lipid nanoparticles modified with fusogenic coiled-coil peptides. NANOSCALE 2023; 15:15206-15218. [PMID: 37671560 DOI: 10.1039/d3nr02175k] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Gene delivery has great potential in modulating protein expression in specific cells to treat diseases. Such therapeutic gene delivery demands sufficient cellular internalization and endosomal escape. Of various nonviral nucleic acid delivery systems, lipid nanoparticles (LNPs) are the most advanced, but still, are very inefficient as the majority are unable to escape from endosomes/lysosomes. Here, we develop a highly efficient gene delivery system using fusogenic coiled-coil peptides. We modified LNPs, carrying EGFP-mRNA, and cells with complementary coiled-coil lipopeptides. Coiled-coil formation between these lipopeptides induced fast nucleic acid uptake and enhanced GFP expression. The cellular uptake of coiled-coil modified LNPs is likely driven by membrane fusion thereby omitting typical endocytosis pathways. This direct cytosolic delivery circumvents the problems commonly observed with the limited endosomal escape of mRNA. Therefore fusogenic coiled-coil peptide modification of existing LNP formulations to enhance nucleic acid delivery efficiency could be beneficial for several gene therapy applications.
Collapse
Affiliation(s)
- Ye Zeng
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Mengjie Shen
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Roy Pattipeiluhu
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Xuequan Zhou
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Yun Zhang
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Thomas Bakkum
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Thomas H Sharp
- Department of Cell and Chemical Biology, Section Electron Microscopy, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Aimee L Boyle
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands.
| |
Collapse
|
16
|
Gori A, Lodigiani G, Colombarolli SG, Bergamaschi G, Vitali A. Cell Penetrating Peptides: Classification, Mechanisms, Methods of Study, and Applications. ChemMedChem 2023; 18:e202300236. [PMID: 37389978 DOI: 10.1002/cmdc.202300236] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
Cell-penetrating peptides (CPPs) encompass a class of peptides that possess the remarkable ability to cross cell membranes and deliver various types of cargoes, including drugs, nucleic acids, and proteins, into cells. For this reason, CPPs are largely investigated in drug delivery applications in the context of many diseases, such as cancer, diabetes, and genetic disorders. While sharing this functionality and some common structural features, such as a high content of positively charged amino acids, CPPs represent an extremely diverse group of elements, which can differentiate under many aspects. In this review, we summarize the most common characteristics of CPPs, introduce their main distinctive features, mechanistic aspects that drive their function, and outline the most widely used techniques for their structural and functional studies. We highlight current gaps and future perspectives in this field, which have the potential to significantly impact the future field of drug delivery and therapeutics.
Collapse
Affiliation(s)
- Alessandro Gori
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Giulia Lodigiani
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Stella G Colombarolli
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, L.go F. Vito 1, 00168, Roma, Italy
| | - Greta Bergamaschi
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Alberto Vitali
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, L.go F. Vito 1, 00168, Roma, Italy
| |
Collapse
|
17
|
Khairkhah N, Namvar A, Bolhassani A. Application of Cell Penetrating Peptides as a Promising Drug Carrier to Combat Viral Infections. Mol Biotechnol 2023; 65:1387-1402. [PMID: 36719639 PMCID: PMC9888354 DOI: 10.1007/s12033-023-00679-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023]
Abstract
Novel effective drugs or therapeutic vaccines have been already developed to eradicate viral infections. Some non-viral carriers have been used for effective drug delivery to a target cell or tissue. Among them, cell penetrating peptides (CPPs) attracted a special interest to enhance drug delivery into the cells with low toxicity. They were also applied to transfer peptide/protein-based and nucleic acids-based therapeutic vaccines against viral infections. CPPs-conjugated drugs or vaccines were investigated in several viral infections including poliovirus, Ebola, coronavirus, herpes simplex virus, human immunodeficiency virus, hepatitis B virus, hepatitis C virus, Japanese encephalitis virus, and influenza A virus. Some studies showed that the uptake of CPPs or CPPs-conjugated drugs can be performed through both non-endocytic and endocytic pathways. Despite high potential of CPPs for cargo delivery, there are some serious drawbacks such as non-tissue-specificity, instability, and suboptimal pharmacokinetics features that limit their clinical applications. At present, some solutions are utilized to improve the CPPs properties such as conjugation of CPPs with targeting moieties, the use of fusogenic lipids, generation of the proton sponge effect, etc. Up to now, no CPP or composition containing CPPs has been approved by the Food and Drug Administration (FDA) due to the lack of sufficient in vivo studies on stability, immunological assays, toxicity, and endosomal escape of CPPs. In this review, we briefly describe the properties, uptake mechanisms, advantages and disadvantages, and improvement of intracellular delivery, and bioavailability of cell penetrating peptides. Moreover, we focus on their application as an effective drug carrier to combat viral infections.
Collapse
Affiliation(s)
- Niloofar Khairkhah
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Namvar
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
18
|
Reveret L, Leclerc M, Morin F, Émond V, Calon F. Pharmacokinetics, biodistribution and toxicology of novel cell-penetrating peptides. Sci Rep 2023; 13:11081. [PMID: 37422520 PMCID: PMC10329699 DOI: 10.1038/s41598-023-37280-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/19/2023] [Indexed: 07/10/2023] Open
Abstract
Cell-penetrating peptides (CPPs) have been used in basic and preclinical research in the past 30 years to facilitate drug delivery into target cells. However, translation toward the clinic has not been successful so far. Here, we studied the pharmacokinetic (PK) and biodistribution profiles of Shuttle cell-penetrating peptides (S-CPP) in rodents, combined or not with an immunoglobulin G (IgG) cargo. We compared two enantiomers of S-CPP that contain both a protein transduction domain and an endosomal escape domain, with previously shown capacity for cytoplasmic delivery. The plasma concentration versus time curve of both radiolabelled S-CPPs required a two-compartment PK analytical model, which showed a fast distribution phase (t1/2α ranging from 1.25 to 3 min) followed by a slower elimination phase (t1/2β ranging from 5 to 15 h) after intravenous injection. Cargo IgG combined to S-CPPs displayed longer elimination half-life, of up to 25 h. The fast decrease in plasma concentration of S-CPPs was associated with an accumulation in target organs assessed at 1 and 5 h post-injection, particularly in the liver. In addition, in situ cerebral perfusion (ISCP) of L-S-CPP yielded a brain uptake coefficient of 7.2 ± 1.1 µl g-1 s-1, consistent with penetration across the blood-brain barrier (BBB), without damaging its integrity in vivo. No sign of peripheral toxicity was detected either by examining hematologic and biochemical blood parameters, or by measuring cytokine levels in plasma. In conclusion, S-CPPs are promising non-toxic transport vectors for improved tissue distribution of drug cargos in vivo.
Collapse
Affiliation(s)
- L Reveret
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Neurosciences Axis, CHU de Québec-Université Laval Research Center, 2705, Boulevard Laurier, Room T2-67, Quebec City, QC, G1V 4G2, Canada
| | - M Leclerc
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Neurosciences Axis, CHU de Québec-Université Laval Research Center, 2705, Boulevard Laurier, Room T2-67, Quebec City, QC, G1V 4G2, Canada
| | - F Morin
- Neurosciences Axis, CHU de Québec-Université Laval Research Center, 2705, Boulevard Laurier, Room T2-67, Quebec City, QC, G1V 4G2, Canada
| | - V Émond
- Neurosciences Axis, CHU de Québec-Université Laval Research Center, 2705, Boulevard Laurier, Room T2-67, Quebec City, QC, G1V 4G2, Canada
| | - F Calon
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada.
- Neurosciences Axis, CHU de Québec-Université Laval Research Center, 2705, Boulevard Laurier, Room T2-67, Quebec City, QC, G1V 4G2, Canada.
| |
Collapse
|
19
|
Anselmo S, Sancataldo G, Baiamonte C, Pizzolanti G, Vetri V. Transportan 10 Induces Perturbation and Pores Formation in Giant Plasma Membrane Vesicles Derived from Cancer Liver Cells. Biomolecules 2023; 13:biom13030492. [PMID: 36979427 PMCID: PMC10046094 DOI: 10.3390/biom13030492] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Continuous progress has been made in the development of new molecules for therapeutic purposes. This is driven by the need to address several challenges such as molecular instability and biocompatibility, difficulties in crossing the plasma membrane, and the development of host resistance. In this context, cell-penetrating peptides (CPPs) constitute a promising tool for the development of new therapies due to their intrinsic ability to deliver therapeutic molecules to cells and tissues. These short peptides have gained increasing attention for applications in drug delivery as well as for their antimicrobial and anticancer activity but the general rules regulating the events involved in cellular uptake and in the following processes are still unclear. Here, we use fluorescence microscopy methods to analyze the interactions between the multifunctional peptide Transportan 10 (TP10) and the giant plasma membrane vesicles (GPMVs) derived from cancer cells. This aims to highlight the molecular mechanisms underlying functional interactions which bring its translocation across the membrane or cytotoxic mechanisms leading to membrane collapse and disruption. The Fluorescence Lifetime Imaging Microscopy (FLIM) method coupled with the phasor approach analysis proved to be the winning choice for following highly dynamic spatially heterogeneous events in real-time and highlighting aspects of such complex phenomena. Thanks to the presented approach, we were able to identify and monitor TP10 translocation into the lumen, internalization, and membrane-induced modifications depending on the peptide concentration regime.
Collapse
Affiliation(s)
- Sara Anselmo
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giuseppe Sancataldo
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Concetta Baiamonte
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giuseppe Pizzolanti
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", Università degli Studi di Palermo, 90127 Palermo, Italy
| | - Valeria Vetri
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
| |
Collapse
|
20
|
Choosing an Optimal Solvent Is Crucial for Obtaining Cell-Penetrating Peptide Nanoparticles with Desired Properties and High Activity in Nucleic Acid Delivery. Pharmaceutics 2023; 15:pharmaceutics15020396. [PMID: 36839718 PMCID: PMC9963036 DOI: 10.3390/pharmaceutics15020396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Cell-penetrating peptides (CPPs) are highly promising transfection agents that can deliver various compounds into living cells, including nucleic acids (NAs). Positively charged CPPs can form non-covalent complexes with negatively charged NAs, enabling simple and time-efficient nanoparticle preparation. However, as CPPs have substantially different chemical and physical properties, their complexation with the cargo and characteristics of the resulting nanoparticles largely depends on the properties of the surrounding environment, i.e., solution. Here, we show that the solvent used for the initial dissolving of a CPP determines the properties of the resulting CPP particles formed in an aqueous solution, including the activity and toxicity of the CPP-NA complexes. Using different biophysical methods such as dynamic light scattering (DLS), atomic force microscopy (AFM), transmission and scanning electron microscopy (TEM and SEM), we show that PepFect14 (PF14), a cationic amphipathic CPP, forms spherical particles of uniform size when dissolved in organic solvents, such as ethanol and DMSO. Water-dissolved PF14, however, tends to form micelles and non-uniform aggregates. When dissolved in organic solvents, PF14 retains its α-helical conformation and biological activity in cell culture conditions without any increase in cytotoxicity. Altogether, our results indicate that by using a solvent that matches the chemical nature of the CPP, the properties of the peptide-cargo particles can be tuned in the desired way. This can be of critical importance for in vivo applications, where CPP particles that are too large, non-uniform, or prone to aggregation may induce severe consequences.
Collapse
|
21
|
Chintapula U, Yang S, Nguyen T, Li Y, Jaworski J, Dong H, Nguyen KT. Supramolecular Peptide Nanofiber/PLGA Nanocomposites for Enhancing Pulmonary Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2022; 14:56498-56509. [PMID: 36475601 DOI: 10.1021/acsami.2c15204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Effective drug delivery to pulmonary sites will benefit from the design and synthesis of novel drug delivery systems that can overcome various tissue and cellular barriers. Cell penetrating peptides (CPPs) have shown promise for intracellular delivery of various imaging probes and therapeutics. Although CPPs improve delivery efficacy to a certain extent, they still lack the scope of engineering to improve the payload capacity and protect the payload from the physiological environment in drug delivery applications. Inspired by recent advances of CPPs and CPP-functionalized nanoparticles, in this work, we demonstrate a novel nanocomposite consisting of fiber-forming supramolecular CPPs that are coated onto polylactic-glycolic acid (PLGA) nanoparticles to enhance pulmonary drug delivery. These nanocomposites show a threefold higher intracellular delivery of nanoparticles in various cells including primary lung epithelial cells, macrophages, and a 10-fold increase in endothelial cells compared to naked PLGA nanoparticles or a twofold increase compared to nanoparticles modified with traditional monomeric CPPs. Cell uptake studies suggest that nanocomposites likely enter cells through mixed macropinocytosis and passive energy-independent mechanisms, which is followed by endosomal escape within 24 h. Nanocomposites also showed potent mucus permeation. More importantly, freeze-drying and nebulizing formulated nanocomposite powder did not affect their physiochemical and biological activity, which further highlights the translative potential for use as a stable drug carrier for pulmonary drug delivery. We expect nanocomposites based on peptide nanofibers, and PLGA nanoparticles can be custom designed to encapsulate and deliver a wide range of therapeutics including nucleic acids, proteins, and small-molecule drugs when employed in inhalable systems to treat various pulmonary diseases.
Collapse
Affiliation(s)
- Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Engineering Research Building, Room 226, 500 UTA Blvd., Arlington, Texas 76010, United States
| | - Su Yang
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Chemistry & Physics Building, Room 130, 700 Planetarium Place, Arlington, Texas 76019, United States
| | - Trinh Nguyen
- Department of Bioengineering, University of Texas at Arlington, Engineering Research Building, Room 226, 500 UTA Blvd., Arlington, Texas 76010, United States
| | - Yang Li
- Department of Biophysics, University of Texas at Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Justyn Jaworski
- Department of Bioengineering, University of Texas at Arlington, Engineering Research Building, Room 226, 500 UTA Blvd., Arlington, Texas 76010, United States
| | - He Dong
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Chemistry & Physics Building, Room 130, 700 Planetarium Place, Arlington, Texas 76019, United States
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Engineering Research Building, Room 226, 500 UTA Blvd., Arlington, Texas 76010, United States
| |
Collapse
|
22
|
Pirhaghi M, Frank SA, Alam P, Nielsen J, Sereikaite V, Gupta A, Strømgaard K, Andreasen M, Sharma D, Saboury AA, Otzen DE. A penetratin-derived peptide reduces the membrane permeabilization and cell toxicity of α-synuclein oligomers. J Biol Chem 2022; 298:102688. [PMID: 36370848 PMCID: PMC9791135 DOI: 10.1016/j.jbc.2022.102688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Parkinson's disease is a neurodegenerative movement disorder associated with the intracellular aggregation of α-synuclein (α-syn). Cytotoxicity is mainly associated with the oligomeric species (αSOs) formed at early stages in α-syn aggregation. Consequently, there is an intense focus on the discovery of novel inhibitors such as peptides to inhibit oligomer formation and toxicity. Here, using peptide arrays, we identified nine peptides with high specificity and affinity for αSOs. Of these, peptides p194, p235, and p249 diverted α-syn aggregation from fibrils to amorphous aggregates with reduced β-structures and increased random coil content. However, they did not reduce αSO cytotoxicity and permeabilization of large anionic unilamellar vesicles. In parallel, we identified a non-self-aggregating peptide (p216), derived from the cell-penetrating peptide penetratin, which showed 12-fold higher binding affinity to αSOs than to α-syn monomers (Kdapp 2.7 and 31.2 μM, respectively). p216 reduced αSOs-induced large anionic unilamellar vesicle membrane permeability at 10-1 to 10-3 mg/ml by almost 100%, was not toxic to SH-SY5Y cells, and reduced αSOs cytotoxicity by about 20%. We conclude that p216 is a promising starting point from which to develop peptides targeting toxic αSOs in Parkinson's disease.
Collapse
Affiliation(s)
- Mitra Pirhaghi
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus C, Denmark; Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Signe Andrea Frank
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus C, Denmark
| | - Parvez Alam
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus C, Denmark; Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Janni Nielsen
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus C, Denmark
| | - Vita Sereikaite
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen Ø, Denmark
| | - Arpit Gupta
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, India
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen Ø, Denmark
| | - Maria Andreasen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Deepak Sharma
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, India; G.N. Ramachandran Protein Centre, Academy of Scientific & Innovative Research, Chennai, India
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - Daniel Erik Otzen
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
23
|
Nguyen Le NM, Zsák S, Le-Vinh B, Friedl JD, Kali G, Knoll P, Seitter HW, Koschak A, Bernkop-Schnürch A. Charge-Converting Nanoemulsions as Promising Retinal Drug and Gene Delivery Systems. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44981-44991. [PMID: 36125912 PMCID: PMC9542710 DOI: 10.1021/acsami.2c11649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/08/2022] [Indexed: 05/31/2023]
Abstract
AIM This study aimed to develop phosphatase-responsive ζ potential converting nanocarriers utilizing polyphosphate-coated cell-penetrating peptide (CPP)-decorated nanoemulsions (NEs) as a novel gene delivery system to retinal cells. METHODS Poly-l-lysine (PLL) was first conjugated with oleylamine (OA) only at its carboxylic end to form the amphiphilic PLL-oleylamine (PLOA) conjugate. Afterward, NEs were loaded with PLOA prior to being coated with tripolyphosphate (TPP) to generate PLOA/TPP NEs. A plasmid containing a reporter gene for green fluorescent protein plasmid (pGFP) was complexed with cationic surfactants forming hydrophobic ion pairs that were loaded in the oily core of NEs. Phosphate removal, ζ potential conversion, and cytotoxicity of the system were evaluated. Cellular uptake and transfection efficiency were investigated in 661W photoreceptor-like cells via microscopic analysis, fluorescence spectroscopy, and flow cytometry. RESULTS Dephosphorylation of PLOA/TPP NEs triggered by alkaline phosphatase (ALP) resulted in the exposure of positive amine groups on the surface of NE droplets and a notable conversion of the ζ potential from -22.4 to +8.5 mV. Cellular uptake of PLOA/TPP NEs performed on 661W photoreceptor-like cells showed a 3-fold increase compared to control NEs. Furthermore, PLOA/TPP NEs also showed low cytotoxicity and high transfection efficacy with ∼50% of cells transfected. CONCLUSIONS Polyphosphate-coated CPP-decorated NEs triggered by ALP could be a promising nanosystem to efficiently deliver drugs and genetic materials to photoreceptor-like cells and other retinal cells for potential treatments of retinal diseases.
Collapse
Affiliation(s)
- Nguyet-Minh Nguyen Le
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Sarah Zsák
- Center
for Chemistry and Biomedicine, Department of Pharmacology and Toxicology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Bao Le-Vinh
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Julian David Friedl
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Gergely Kali
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Patrick Knoll
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Hartwig Wolfram Seitter
- Center
for Chemistry and Biomedicine, Department of Pharmacology and Toxicology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Alexandra Koschak
- Center
for Chemistry and Biomedicine, Department of Pharmacology and Toxicology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Center
for Chemistry and Biomedicine, Department of Pharmaceutical Technology,
Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| |
Collapse
|
24
|
Parrasia S, Szabò I, Zoratti M, Biasutto L. Peptides as Pharmacological Carriers to the Brain: Promises, Shortcomings and Challenges. Mol Pharm 2022; 19:3700-3729. [PMID: 36174227 DOI: 10.1021/acs.molpharmaceut.2c00523] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Central nervous system (CNS) diseases are among the most difficult to treat, mainly because the vast majority of the drugs fail to cross the blood-brain barrier (BBB) or to reach the brain at concentrations adequate to exert a pharmacological activity. The obstacle posed by the BBB has led to the in-depth study of strategies allowing the brain delivery of CNS-active drugs. Among the most promising strategies is the use of peptides addressed to the BBB. Peptides are versatile molecules that can be used to decorate nanoparticles or can be conjugated to drugs, with either a stable link or as pro-drugs. They have been used to deliver to the brain both small molecules and proteins, with applications in diverse therapeutic areas such as brain cancers, neurodegenerative diseases and imaging. Peptides can be generally classified as receptor-targeted, recognizing membrane proteins expressed by the BBB microvessels (e.g., Angiopep2, CDX, and iRGD), "cell-penetrating peptides" (CPPs; e.g. TAT47-57, SynB1/3, and Penetratin), undergoing transcytosis through unspecific mechanisms, or those exploiting a mixed approach. The advantages of peptides have been extensively pointed out, but so far few studies have focused on the potential negative aspects. Indeed, despite having a generally good safety profile, some peptide conjugates may display toxicological characteristics distinct from those of the peptide itself, causing for instance antigenicity, cardiovascular alterations or hemolysis. Other shortcomings are the often brief lifetime in vivo, caused by the presence of peptidases, the vulnerability to endosomal/lysosomal degradation, and the frequently still insufficient attainable increase of brain drug levels, which remain below the therapeutically useful concentrations. The aim of this review is to analyze not only the successful and promising aspects of the use of peptides in brain targeting but also the problems posed by this strategy for drug delivery.
Collapse
Affiliation(s)
- Sofia Parrasia
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Mario Zoratti
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Lucia Biasutto
- CNR Neuroscience Institute, Viale G. Colombo 3, 35131 Padova, Italy.,Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| |
Collapse
|
25
|
Chablani L, Singh V. Cell-Penetrating Peptides as Passive Permeation Enhancers for Transdermal Drug Delivery. AAPS PharmSciTech 2022; 23:266. [PMID: 36163537 DOI: 10.1208/s12249-022-02424-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/15/2022] [Indexed: 11/30/2022] Open
Abstract
Cell-penetrating peptides have been widely used as a tool to gain access to cytosol for numerous applications. The review highlights the advances made in preclinical and clinical research using cell-penetrating peptides since their discovery in 1980s. Further, the emphasis is on summarizing the role of cell-penetrating peptides as permeation enhancers for transdermal and topical drug delivery applications. A summary table of preclinical studies utilizing various peptides in combination with different active ingredients and drug delivery systems is included. Lastly, we capture the challenges associated with the cell-penetrating peptides to translate the preclinical work to clinical applications.
Collapse
Affiliation(s)
- Lipika Chablani
- Department of Pharmaceutical Sciences, Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, New York, 14618, USA.
| | - Vijay Singh
- Research and Development, Bausch and Lomb, 1400 North Goodman Street, Rochester, New York, 14609, USA
| |
Collapse
|
26
|
Lou J, Qualls ML, Hudson MM, McBee DP, Baccile JA, Best MD. Reactive Oxygen Species (ROS) Activated Liposomal Cell Delivery using a Boronate-Caged Guanidine Lipid. Chemistry 2022; 28:e202201057. [PMID: 35639353 PMCID: PMC9388614 DOI: 10.1002/chem.202201057] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Indexed: 08/18/2023]
Abstract
We report boronate-caged guanidine-lipid 1 that activates liposomes for cellular delivery only upon uncaging of this compound by reactive oxygen species (ROS) to produce cationic lipid products. These liposomes are designed to mimic the exceptional cell delivery properties of cell-penetrating peptides (CPPs), while the inclusion of the boronate cage is designed to enhance selectivity such that cell entry will only be activated in the presence of ROS. Boronate uncaging by hydrogen peroxide was verified by mass spectrometry and zeta potential (ZP) measurements. A microplate-based fluorescence assay was developed to study the ROS-mediated vesicle interactions between 1-liposomes and anionic membranes, which were further elucidated via dynamic light scattering (DLS) analysis. Cellular delivery studies utilizing fluorescence microscopy demonstrated significant enhancements in cellular delivery only when 1-liposomes were incubated with hydrogen peroxide. Our results showcase that lipid 1 exhibits strong potential as an ROS-responsive liposomal platform for targeted drug delivery applications.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Megan L Qualls
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Macy M Hudson
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Dillon P McBee
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Joshua A Baccile
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| |
Collapse
|
27
|
Silva S, Kurrikoff K, Langel Ü, Almeida AJ, Vale N. A Second Life for MAP, a Model Amphipathic Peptide. Int J Mol Sci 2022; 23:8322. [PMID: 35955457 PMCID: PMC9368858 DOI: 10.3390/ijms23158322] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/26/2022] Open
Abstract
Cell-penetrating peptides (CPP) have been shown to be efficient in the transport of cargoes into the cells, namely siRNA and DNA, proteins and peptides, and in some cases, small therapeutics. These peptides have emerged as a solution to increase drug concentrations in different tissues and various cell types, therefore having a relevant therapeutic relevance which led to clinical trials. One of them, MAP, is a model amphipathic peptide with an α-helical conformation and both hydrophilic and hydrophobic residues in opposite sides of the helix. It is composed of a mixture of alanines, leucines, and lysines (KLALKLALKALKAALKLA). The CPP MAP has the ability to translocate oligonucleotides, peptides and small proteins. However, taking advantage of its unique properties, in recent years innovative concepts were developed, such as in silico studies of modelling with receptors, coupling and repurposing drugs in the central nervous system and oncology, or involving the construction of dual-drug delivery systems using nanoparticles. In addition to designs of MAP-linked vehicles and strategies to achieve highly effective yet less toxic chemotherapy, this review will be focused on unique molecular structure and how it determines its cellular activity, and also intends to address the most recent and frankly motivating issues for the future.
Collapse
Affiliation(s)
- Sara Silva
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal;
| | - Kaido Kurrikoff
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia; (K.K.); (Ü.L.)
| | - Ülo Langel
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia; (K.K.); (Ü.L.)
- Department of Biochemistry and Biophysics, Stockholm University, 10691 Stockholm, Sweden
| | - António J. Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal;
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
28
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Naffouje SA, Goto M, Coward LU, Gorman GS, Christov K, Wang J, Green A, Shilkaitis A, Das Gupta TK, Yamada T. Nontoxic Tumor-Targeting Optical Agents for Intraoperative Breast Tumor Imaging. J Med Chem 2022; 65:7371-7379. [PMID: 35544687 DOI: 10.1021/acs.jmedchem.2c00417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Precise identification of the tumor margins during breast-conserving surgery (BCS) remains a challenge given the lack of visual discrepancy between malignant and surrounding normal tissues. Therefore, we developed a fluorescent imaging agent, ICG-p28, for intraoperative imaging guidance to better aid surgeons in achieving negative margins in BCS. Here, we determined the pharmacokinetics (PK), biodistribution, and preclinical toxicity of ICG-p28. The PK and biodistribution of ICG-p28 indicated rapid tissue uptake and localization at tumor lesions. There were no dose-related effect and no significant toxicity in any of the breast cancer and normal cell lines tested. Furthermore, ICG-p28 was evaluated in clinically relevant settings with transgenic mice that spontaneously developed invasive mammary tumors. Intraoperative imaging with ICG-p28 showed a significant reduction in the tumor recurrence rate. This simple, nontoxic, and cost-effective method can offer a new approach that enables surgeons to intraoperatively identify tumor margins and potentially improves overall outcomes by reducing recurrence rates.
Collapse
Affiliation(s)
- Samer A Naffouje
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Masahide Goto
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Lori U Coward
- McWhorter School of Pharmacy, Pharmaceutical, Social and Administrative Sciences, Samford University, Birmingham, Alabama 35229, United States
| | - Gregory S Gorman
- McWhorter School of Pharmacy, Pharmaceutical, Social and Administrative Sciences, Samford University, Birmingham, Alabama 35229, United States
| | - Konstantin Christov
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Jing Wang
- Department of Mathematics, Statistics and Computer Science, University of Illinois College of Liberal Arts and Sciences, Urbana, Illinois 60612, United States
| | - Albert Green
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Anne Shilkaitis
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Tapas K Das Gupta
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States.,Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Medicine and Engineering, Chicago, Illinois 60607, United States
| |
Collapse
|
30
|
Paquette AR, Payne SR, McKay GA, Brazeau-Henrie JT, Darnowski MG, Kammili A, Bernal F, Mah TF, Gruenheid S, Nguyen D, Boddy CN. RpoN-Based stapled peptides with improved DNA binding suppress Pseudomonas aeruginosa virulence. RSC Med Chem 2022; 13:445-455. [PMID: 35647551 PMCID: PMC9020619 DOI: 10.1039/d1md00371b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/02/2022] [Indexed: 11/21/2022] Open
Abstract
Stapled peptides have the ability to mimic α-helices involved in protein binding and have proved to be effective pharmacological agents for disrupting protein-protein interactions. DNA-binding proteins such as transcription factors bind their cognate DNA sequences via an α-helix interacting with the major groove of DNA. We previously developed a stapled peptide based on the bacterial alternative sigma factor RpoN capable of binding the RpoN DNA promoter sequence and inhibiting RpoN-mediated expression in Escherichia coli. We have elucidated a structure-activity relationship for DNA binding by this stapled peptide, improving DNA binding affinity constants in the high nM range. Lead peptides were shown to have low toxicity as determined by their low hemolytic activity at 100 μM and were shown to have anti-virulence activity in a Galleria mellonella model of Pseudomonas aeruginosa infection. These findings support further preclinical development of stapled peptides as antivirulence agents targeting P. aeruginosa.
Collapse
Affiliation(s)
- André R. Paquette
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaONK1N 6N5 Canada
| | - Sterling R. Payne
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, National Institutes of HealthFrederickMD 21702USA
| | - Geoffrey A. McKay
- Meakins-Christie Laboratories, Research Institute of the McGill University Health CentreMontrealQuebec H4A 3J1Canada
| | | | - Micheal G. Darnowski
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaONK1N 6N5 Canada
| | - Anitha Kammili
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa ON K1N 6N5 Canada
| | - Federico Bernal
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, National Institutes of HealthFrederickMD 21702USA
| | - Thien-Fah Mah
- Department of Biochemistry, Microbiology, and Immunology, University of OttawaOttawaONK1H 8M5Canada
| | | | - Dao Nguyen
- Meakins-Christie Laboratories, Research Institute of the McGill University Health CentreMontrealQuebec H4A 3J1Canada,Department of Medicine, McGill UniversityMontrealQuebec H4A 3J1Canada
| | - Christopher N. Boddy
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaONK1N 6N5 Canada
| |
Collapse
|
31
|
Choi H, Choi K, Kim DH, Oh BK, Yim H, Jo S, Choi C. Strategies for Targeted Delivery of Exosomes to the Brain: Advantages and Challenges. Pharmaceutics 2022; 14:672. [PMID: 35336049 PMCID: PMC8948948 DOI: 10.3390/pharmaceutics14030672] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/08/2023] Open
Abstract
Delivering therapeutics to the central nervous system (CNS) is difficult because of the blood-brain barrier (BBB). Therapeutic delivery across the tight junctions of the BBB can be achieved through various endogenous transportation mechanisms. Receptor-mediated transcytosis (RMT) is one of the most widely investigated and used methods. Drugs can hijack RMT by expressing specific ligands that bind to receptors mediating transcytosis, such as the transferrin receptor (TfR), low-density lipoprotein receptor (LDLR), and insulin receptor (INSR). Cell-penetrating peptides and viral components originating from neurotropic viruses can also be utilized for the efficient BBB crossing of therapeutics. Exosomes, or small extracellular vesicles, have gained attention as natural nanoparticles for treating CNS diseases, owing to their potential for natural BBB crossing and broad surface engineering capability. RMT-mediated transport of exosomes expressing ligands such as LDLR-targeting apolipoprotein B has shown promising results. Although surface-modified exosomes possessing brain targetability have shown enhanced CNS delivery in preclinical studies, the successful development of clinically approved exosome therapeutics for CNS diseases requires the establishment of quantitative and qualitative methods for monitoring exosomal delivery to the brain parenchyma in vivo as well as elucidation of the mechanisms underlying the BBB crossing of surface-modified exosomes.
Collapse
Affiliation(s)
- Hojun Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Kyungsun Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Dae-Hwan Kim
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Byung-Koo Oh
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Hwayoung Yim
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Soojin Jo
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (K.C.); (D.-H.K.); (B.-K.O.); (H.Y.); (S.J.)
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
32
|
Pnueli L, Melamed P. Epigenetic repression of gonadotropin gene expression via a GnRH-mediated DNA delivery system. Gene Ther 2022; 29:294-303. [PMID: 35301447 DOI: 10.1038/s41434-022-00325-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 02/08/2022] [Accepted: 02/18/2022] [Indexed: 11/09/2022]
Abstract
The reproductive axis is activated by gonadotropin-releasing hormone (GnRH), which stimulates the pituitary gonadotropes to secrete hormones that drive gonadal function and steroidogenesis. Thus repression of this axis, which is conserved across mammals and sexes, can reduce steroid levels and/or prevent reproduction. Steroid-dependent pathologies, including various cancers, are commonly treated with GnRH super-analogs which have long-term side-effects, while humane solutions for controlling reproduction in domestic and wild animal populations are lacking. GnRH-conjugated toxins are undergoing clinical trials for GnRHR-expressing cancer cells, and have been examined for gonadotrope ablation in animals, but showed low and/or transient effects and administration of toxins has many potential complications. Here we exploit GnRH targeting to gonadotropes to deliver DNA encoding an effector that induces gonadotropin gene repressive epigenetic modifications which are perpetuated over time. Several layers of specificity are endowed through targeting to GnRHR-expressing cells and due to local cleavage of the peptide packaging the DNA; the DNA-encoded effector is expressed and directed to the target genes by the DNA binding domain of a highly specific transcription factor. This design has multiple advantages over existing methods of shutting down the reproductive axis, and its modular design should allow adaptation for broad applications.
Collapse
Affiliation(s)
- Lilach Pnueli
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel
| | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| |
Collapse
|
33
|
Robles-Loaiza AA, Pinos-Tamayo EA, Mendes B, Ortega-Pila JA, Proaño-Bolaños C, Plisson F, Teixeira C, Gomes P, Almeida JR. Traditional and Computational Screening of Non-Toxic Peptides and Approaches to Improving Selectivity. Pharmaceuticals (Basel) 2022; 15:323. [PMID: 35337121 PMCID: PMC8953747 DOI: 10.3390/ph15030323] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/27/2022] Open
Abstract
Peptides have positively impacted the pharmaceutical industry as drugs, biomarkers, or diagnostic tools of high therapeutic value. However, only a handful have progressed to the market. Toxicity is one of the main obstacles to translating peptides into clinics. Hemolysis or hemotoxicity, the principal source of toxicity, is a natural or disease-induced event leading to the death of vital red blood cells. Initial screenings for toxicity have been widely evaluated using erythrocytes as the gold standard. More recently, many online databases filled with peptide sequences and their biological meta-data have paved the way toward hemolysis prediction using user-friendly, fast-access machine learning-driven programs. This review details the growing contributions of in silico approaches developed in the last decade for the large-scale prediction of erythrocyte lysis induced by peptides. After an overview of the pharmaceutical landscape of peptide therapeutics, we highlighted the relevance of early hemolysis studies in drug development. We emphasized the computational models and algorithms used to this end in light of historical and recent findings in this promising field. We benchmarked seven predictors using peptides from different data sets, having 7-35 amino acids in length. According to our predictions, the models have scored an accuracy over 50.42% and a minimal Matthew's correlation coefficient over 0.11. The maximum values for these statistical parameters achieved 100.0% and 1.00, respectively. Finally, strategies for optimizing peptide selectivity were described, as well as prospects for future investigations. The development of in silico predictive approaches to peptide toxicity has just started, but their important contributions clearly demonstrate their potential for peptide science and computer-aided drug design. Methodology refinement and increasing use will motivate the timely and accurate in silico identification of selective, non-toxic peptide therapeutics.
Collapse
Affiliation(s)
- Alberto A. Robles-Loaiza
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Tena 150150, Ecuador; (A.A.R.-L.); (B.M.); (J.A.O.-P.); (C.P.-B.)
| | - Edgar A. Pinos-Tamayo
- Escuela Superior Politécnica del Litoral, ESPOL, Centro Nacional de Acuicultura e Investigaciones Marinas (CENAIM), Campus Gustavo Galindo Km. 30, 5 Vía Perimetral, Guayaquil 09-01-5863, Ecuador;
| | - Bruno Mendes
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Tena 150150, Ecuador; (A.A.R.-L.); (B.M.); (J.A.O.-P.); (C.P.-B.)
| | - Josselyn A. Ortega-Pila
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Tena 150150, Ecuador; (A.A.R.-L.); (B.M.); (J.A.O.-P.); (C.P.-B.)
| | - Carolina Proaño-Bolaños
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Tena 150150, Ecuador; (A.A.R.-L.); (B.M.); (J.A.O.-P.); (C.P.-B.)
| | - Fabien Plisson
- Consejo Nacional de Ciencia y Tecnología, Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Centro de Investigación Y de Estudios Avanzados del IPN, Irapuato 36824, Mexico;
| | - Cátia Teixeira
- Laboratório Associado para a Química Verde-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal; (C.T.); (P.G.)
| | - Paula Gomes
- Laboratório Associado para a Química Verde-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal; (C.T.); (P.G.)
| | - José R. Almeida
- Biomolecules Discovery Group, Universidad Regional Amazónica Ikiam, Tena 150150, Ecuador; (A.A.R.-L.); (B.M.); (J.A.O.-P.); (C.P.-B.)
| |
Collapse
|
34
|
Samec T, Boulos J, Gilmore S, Hazelton A, Alexander-Bryant A. Peptide-based delivery of therapeutics in cancer treatment. Mater Today Bio 2022; 14:100248. [PMID: 35434595 PMCID: PMC9010702 DOI: 10.1016/j.mtbio.2022.100248] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Abstract
Current delivery strategies for cancer therapeutics commonly cause significant systemic side effects due to required high doses of therapeutic, inefficient cellular uptake of drug, and poor cell selectivity. Peptide-based delivery systems have shown the ability to alleviate these issues and can significantly enhance therapeutic loading, delivery, and cancer targetability. Peptide systems can be tailor-made for specific cancer applications. This review describes three peptide classes, targeting, cell penetrating, and fusogenic peptides, as stand-alone nanoparticle systems, conjugations to nanoparticle systems, or as the therapeutic modality. Peptide nanoparticle design, characteristics, and applications are discussed as well as peptide applications in the clinical space.
Collapse
Affiliation(s)
- Timothy Samec
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Jessica Boulos
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Serena Gilmore
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Anthony Hazelton
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Angela Alexander-Bryant
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| |
Collapse
|
35
|
Rahman A, Matthews MA, Nowell CJ, Chalmers DK, Thompson PE, Nicholson SE, Barlow N, Norton RS. Enhanced nitric oxide production by macrophages treated with a cell-penetrating peptide conjugate. Bioorg Chem 2022; 123:105763. [DOI: 10.1016/j.bioorg.2022.105763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/02/2022]
|
36
|
Wang XL, Jiang RW. Therapeutic Potential of Superoxide Dismutase Fused with Cell-Penetrating Peptides in Oxidative Stress-Related Diseases. Mini Rev Med Chem 2022; 22:2287-2298. [PMID: 35227183 DOI: 10.2174/1389557522666220228150127] [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: 11/26/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 11/22/2022]
Abstract
Superoxide dismutase (SOD) is a well-known cellular antioxidant enzyme. However, exogenous SOD cannot be used to protect tissues from oxidative damage due to the low permeability of the cell membrane. Cell-penetrating peptides (CPPs) are a class of short peptides that can cross the cell membrane. Recombinant fusion protein that fuses SOD protein with CPP (CPP-SOD) can cross various tissues and organs as well as the blood-brain barrier. CPP-SODs can relieve severe oxidative damage in various tissues caused by radiation, ischemia, inflammation, and chemotherapy by clearing the reactive oxygen species, reducing the expression of inflammatory factors, and inhibiting NF-κB/MAPK signaling pathways. Therefore, the clinical application of CPP-SODs provide new therapeutic strategies for a variety of oxidative stress-related disorders, such as Parkinson's disease, diabetes, obesity, cardiac fibrosis, and premature aging.
Collapse
Affiliation(s)
- Xiao-Lu Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University, Guangzhou 510632, China
| |
Collapse
|
37
|
Maloverjan M, Padari K, Abroi A, Rebane A, Pooga M. Divalent Metal Ions Boost Effect of Nucleic Acids Delivered by Cell-Penetrating Peptides. Cells 2022; 11:cells11040756. [PMID: 35203400 PMCID: PMC8870069 DOI: 10.3390/cells11040756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/11/2022] [Accepted: 02/19/2022] [Indexed: 12/01/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are promising tools for the transfection of various substances, including nucleic acids, into cells. The aim of the current work was to search for novel safe and effective approaches for enhancing transfection efficiency of nanoparticles formed from CPP and splice-correcting oligonucleotide (SCO) without increasing the concentration of peptide. We analyzed the effect of inclusion of calcium and magnesium ions into nanoparticles on CPP-mediated transfection in cell culture. We also studied the mechanism of such transfection as well as its efficiency, applicability in case of different cell lines, nucleic acid types and peptides, and possible limitations. We discovered a strong positive effect of these ions on transfection efficiency of SCO, that translated to enhanced synthesis of functional reporter protein. We observed significant changes in intracellular distribution and trafficking of nanoparticles formed by the addition of the ions, without increasing cytotoxicity. We propose a novel strategy for preparing CPP-oligonucleotide nanoparticles with enhanced efficiency and, thus, higher therapeutic potential. Our discovery may be translated to primary cell cultures and, possibly, in vivo studies, with the aim of increasing CPP-mediated transfection efficiency and the likelihood of using CPPs in clinics.
Collapse
Affiliation(s)
- Maria Maloverjan
- Institute of Technology, University of Tartu, 1 Nooruse Street, 50411 Tartu, Estonia; (M.M.); (A.A.)
| | - Kärt Padari
- Institute of Molecular and Cell Biology, University of Tartu, 23b Riia Street, 51010 Tartu, Estonia;
| | - Aare Abroi
- Institute of Technology, University of Tartu, 1 Nooruse Street, 50411 Tartu, Estonia; (M.M.); (A.A.)
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine, University of Tartu, 14b Ravila Street, 50411 Tartu, Estonia;
| | - Margus Pooga
- Institute of Technology, University of Tartu, 1 Nooruse Street, 50411 Tartu, Estonia; (M.M.); (A.A.)
- Correspondence: ; Tel.: +372-737-4836
| |
Collapse
|
38
|
Egorova A, Shtykalova S, Maretina M, Selutin A, Shved N, Deviatkin D, Selkov S, Baranov V, Kiselev A. Polycondensed Peptide Carriers Modified with Cyclic RGD Ligand for Targeted Suicide Gene Delivery to Uterine Fibroid Cells. Int J Mol Sci 2022; 23:1164. [PMID: 35163086 PMCID: PMC8835468 DOI: 10.3390/ijms23031164] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Suicide gene therapy was suggested as a possible strategy for the treatment of uterine fibroids (UFs), which are the most common benign tumors inwomen of reproductive age. For successful suicide gene therapy, DNAtherapeutics should be specifically delivered to UF cells. Peptide carriers are promising non-viral gene delivery systems that can be easily modified with ligands and other biomolecules to overcome DNA transfer barriers. Here we designed polycondensed peptide carriers modified with a cyclic RGD moiety for targeted DNA delivery to UF cells. Molecular weights of the resultant polymers were determined, and inclusion of the ligand was confirmed by MALDI-TOF. The physicochemical properties of the polyplexes, as well as cellular DNA transport, toxicity, and transfection efficiency were studied, and the specificity of αvβ3 integrin-expressing cell transfection was proved. The modification with the ligand resulted in a three-fold increase of transfection efficiency. Modeling of the suicide gene therapy by transferring the HSV-TK suicide gene to primary cells obtained from myomatous nodes of uterine leiomyoma patients was carried out. We observed up to a 2.3-fold decrease in proliferative activity after ganciclovir treatment of the transfected cells. Pro- and anti-apoptotic gene expression analysis confirmed our findings that the developed polyplexes stimulate UF cell death in a suicide-specific manner.
Collapse
Affiliation(s)
- Anna Egorova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Sofia Shtykalova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Marianna Maretina
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Alexander Selutin
- Department of Immunology and Intercellular Interactions, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.S.); (S.S.)
| | - Natalia Shved
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Dmitriy Deviatkin
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Sergey Selkov
- Department of Immunology and Intercellular Interactions, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.S.); (S.S.)
| | - Vladislav Baranov
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| | - Anton Kiselev
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (A.E.); (S.S.); (M.M.); (N.S.); (D.D.); (V.B.)
| |
Collapse
|
39
|
Systematic Screening of Penetratin's Protein Targets by Yeast Proteome Microarrays. Int J Mol Sci 2022; 23:ijms23020712. [PMID: 35054898 PMCID: PMC8775591 DOI: 10.3390/ijms23020712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Cell-penetrating peptides (CPPs) have distinct properties to translocate across cell envelope. The key property of CPPs to translocation with attached molecules has been utilized as vehicles for the delivery of several potential drug candidates that illustrate the significant effect in in-vitro experiment but fail in in-vivo experiment due to selectively permeable nature of cell envelop. Penetratin, a well-known CPP identified from the third α-helix of Antennapedia homeodomain of Drosophila, has been widely used and studied for the delivery of bioactive molecules to treat cancers, stroke, and infections caused by pathogenic organisms. Few studies have demonstrated that penetratin directly possesses antimicrobial activities against bacterial and fungal pathogens; however, the mechanism is unknown. In this study, we have utilized the power of high-throughput Saccharomyces cerevisiae proteome microarrays to screen all the potential protein targets of penetratin. Saccharomyces cerevisiae proteome microarrays assays of penetratin followed by statistical analysis depicted 123 Saccharomyces cerevisiae proteins as the protein targets of penetratin out of ~5800 Saccharomyces cerevisiae proteins. To understand the target patterns of penetratin, enrichment analyses were conducted using 123 protein targets. In biological process: ribonucleoprotein complex biogenesis, nucleic acid metabolic process, actin filament-based process, transcription, DNA-templated, and negative regulation of gene expression are a few significantly enriched terms. Cytoplasm, nucleus, and cell-organelles are enriched terms for cellular component. Protein-protein interactions network depicted ribonucleoprotein complex biogenesis, cortical cytoskeleton, and histone binding, which represent the major enriched terms for the 123 protein targets of penetratin. We also compared the protein targets of penetratin and intracellular protein targets of antifungal AMPs (Lfcin B, Histatin-5, and Sub-5). The comparison results showed few unique proteins between penetratin and AMPs. Nucleic acid metabolic process and cellular component disassembly were the common enrichment terms for penetratin and three AMPs. Penetratin shows unique enrichment items that are related to DNA biological process. Moreover, motif enrichment analysis depicted different enriched motifs in the protein targets of penetratin, LfcinB, Histatin-5, and Sub-5.
Collapse
|
40
|
Farooq Z, Howell LA, McCormick PJ. Probing GPCR Dimerization Using Peptides. Front Endocrinol (Lausanne) 2022; 13:843770. [PMID: 35909575 PMCID: PMC9329873 DOI: 10.3389/fendo.2022.843770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest class of membrane proteins and the most common and extensively studied pharmacological target. Numerous studies over the last decade have confirmed that GPCRs do not only exist and function in their monomeric form but in fact, have the ability to form dimers or higher order oligomers with other GPCRs, as well as other classes of receptors. GPCR oligomers have become increasingly attractive to investigate as they have the ability to modulate the pharmacological responses of the receptors which in turn, could have important functional roles in diseases, such as cancer and several neurological & neuropsychiatric disorders. Despite the growing evidence in the field of GPCR oligomerisation, the lack of structural information, as well as targeting the 'undruggable' protein-protein interactions (PPIs) involved in these complexes, has presented difficulties. Outside the field of GPCRs, targeting PPIs has been widely studied, with a variety of techniques being investigated; from small-molecule inhibitors to disrupting peptides. In this review, we will demonstrate several physiologically relevant GPCR dimers and discuss an array of strategies and techniques that can be employed when targeting these complexes, as well as provide ideas for future development.
Collapse
Affiliation(s)
- Zara Farooq
- Centre for Endocrinology, William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, Mile End Road, London, United Kingdom
| | - Lesley A. Howell
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, Mile End Road, London, United Kingdom
| | - Peter J. McCormick
- Centre for Endocrinology, William Harvey Research Institute, Bart’s and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- *Correspondence: Peter J. McCormick,
| |
Collapse
|
41
|
Abstract
In this introductory chapter, we first define cell-penetrating peptides (CPPs), give short overview of CPP history and discuss several aspects of CPP classification. Next section is devoted to the mechanism of CPP penetration into the cells, where direct and endocytic internalization of CPP is explained. Kinetics of internalization is discussed more extensively, since this topic is not discussed in other chapters of this book. At the end of this section some features of the thermodynamics of CPP interaction with the membrane is also presented. Finally, we present different cargoes that can be transferred into the cells by CPPs and briefly discuss the effect of cargo on the rate and efficiency of penetration into the cells.
Collapse
Affiliation(s)
- Matjaž Zorko
- Medical Faculty, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia.
| | - Ülo Langel
- Department of Biochemistry and Biophysics, University of Stockholm, Stockholm, Sweden.,Institute of Technology, University of Tartu, Tartu, Estonia
| |
Collapse
|
42
|
Synthetic Molecular Evolution of Cell Penetrating Peptides. Methods Mol Biol 2021; 2383:73-89. [PMID: 34766283 DOI: 10.1007/978-1-0716-1752-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Rational design and optimization of cell penetrating peptides (CPPs) is difficult to accomplish because of the lack of quantitative sequence-structure-function rules describing the activity and because of the complex, poorly understood mechanisms of CPPs. Synthetic molecular evolution is a powerful method to identify gain-of-function cell penetrating peptide variants in this situation. Synthetic molecular evolution requires the design and synthesis of iterative, knowledge-based peptide libraries and the screening of such libraries in complex orthogonal cell-based screens for improved activity. In this chapter, we describe methods for synthesizing powerful combinatorial peptide libraries for synthetic molecular evolution.
Collapse
|
43
|
Anselmo S, Sancataldo G, Mørck Nielsen H, Foderà V, Vetri V. Peptide-Membrane Interactions Monitored by Fluorescence Lifetime Imaging: A Study Case of Transportan 10. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:13148-13159. [PMID: 34714654 PMCID: PMC8582253 DOI: 10.1021/acs.langmuir.1c02392] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/19/2021] [Indexed: 06/13/2023]
Abstract
The interest on detailed analysis of peptide-membrane interactions is of great interest in both fundamental and applied sciences as these may relate to both functional and pathogenic events. Such interactions are highly dynamic and spatially heterogeneous, making the investigation of the associated phenomena highly complex. The specific properties of membranes and peptide structural details, together with environmental conditions, may determine different events at the membrane interface, which will drive the fate of the peptide-membrane system. Here, we use an experimental approach based on the combination of spectroscopy and fluorescence microscopy methods to characterize the interactions of the multifunctional amphiphilic peptide transportan 10 with model membranes. Our approach, based on the use of suitable fluorescence reporters, exploits the advantages of phasor plot analysis of fluorescence lifetime imaging microscopy measurements to highlight the molecular details of occurring membrane alterations in terms of rigidity and hydration. Simultaneously, it allows following dynamic events in real time without sample manipulation distinguishing, with high spatial resolution, whether the peptide is adsorbed to or inserted in the membrane.
Collapse
Affiliation(s)
- Sara Anselmo
- Dipartimento
di Fisica e Chimica−Emilio Segré, Università degli Studi di Palermo, Viale delle Scienze ed. 18 90128, Palermo, Italy
| | - Giuseppe Sancataldo
- Dipartimento
di Fisica e Chimica−Emilio Segré, Università degli Studi di Palermo, Viale delle Scienze ed. 18 90128, Palermo, Italy
| | - Hanne Mørck Nielsen
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2 2100, Copenhagen, Denmark
| | - Vito Foderà
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2 2100, Copenhagen, Denmark
| | - Valeria Vetri
- Dipartimento
di Fisica e Chimica−Emilio Segré, Università degli Studi di Palermo, Viale delle Scienze ed. 18 90128, Palermo, Italy
| |
Collapse
|
44
|
Ong SY, Zhang C, Dong X, Yao SQ. Recent Advances in Polymeric Nanoparticles for Enhanced Fluorescence and Photoacoustic Imaging. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Sing Yee Ong
- Department of Chemistry National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- National University of Singapore Graduate School (Integrative Sciences and Engineering Programme, ISEP) National University of Singapore University Hall, Tan Chin Tuan Wing, 21 Lower Kent Ridge Road, #04-02 Singapore 119077 Singapore
| | - Changyu Zhang
- Department of Chemistry National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Xiao Dong
- Department of Chemistry National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Shao Q. Yao
- Department of Chemistry National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- National University of Singapore Graduate School (Integrative Sciences and Engineering Programme, ISEP) National University of Singapore University Hall, Tan Chin Tuan Wing, 21 Lower Kent Ridge Road, #04-02 Singapore 119077 Singapore
| |
Collapse
|
45
|
Zhang Z, Ma W, He K, Yuan B, Yang K. Ligand-decoration determines the translational and rotational dynamics of nanoparticles on a lipid bilayer membrane. Phys Chem Chem Phys 2021; 23:9158-9165. [PMID: 33885120 DOI: 10.1039/d1cp00643f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanoparticles (NPs) promise a huge potential for clinical diagnostic and therapeutic applications. However, nano-bio (e.g., the NP-cell membrane) interactions and underlying mechanisms are still largely elusive. In this study, two types of congeneric peptides, namely PGLa and magainin 2 (MAG2), with similar membrane activities were employed as model ligands for NP decoration, and the diffusion behaviours (including both translation and rotation) of the ligand-decorated NPs on a lipid bilayer membrane were studied via molecular dynamics simulations. It was found that, although both PGLa- and MAG2-coated NPs showed alternatively "hopping" and "jiggling" diffusions, the PGLa-coated ones had an enhanced circling at the hopping stage, while a much confined circling at the jiggling stage. In contrast, the MAG2-coated NPs demonstrated constant circling tendencies throughout the diffusion process. Such differences in the coupling between translational and rotational dynamics of these two types of NPs are ascribed to the different ligand-lipid interactions of PGLa and MAG2, in which the PGLa ligands prefer to vertically insert into the membrane, while MAG2 tends to lie flat on the membrane surface. Our results are helpful for the understanding the underlying associations between the NP motions and their interfacial membrane interactions, and shed light on the possibility of regulating NP behaviours on a cellular surface for better biomedical uses.
Collapse
Affiliation(s)
- Zhihong Zhang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, 215006, P. R. China.
| | | | | | | | | |
Collapse
|
46
|
Engineered nanoplex mediated targeted miRNA delivery to rescue dying podocytes in diabetic nephropathy. Int J Pharm 2021; 605:120842. [PMID: 34216766 DOI: 10.1016/j.ijpharm.2021.120842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/08/2023]
Abstract
MicroRNAs (miRNA) is vital for gene expression regulation and normal kidney function. Mainly, miRNA-30a is responsible for the homeostasis of podocytes. In the diabetic nephropathic condition, miRNA-30a is directly and primarily suppressed by hyperglycemic kidney induced Notch signaling pathway leads to podocyte damage and apoptosis. Thus, transferring the exogenous miRNA-30a to podocytes might improve albuminuria as well as podocytes injury. The deprived stability, poor targetability, and low specificity in vivo are critical limitations to attain this objective. This investigation reports the specific and efficient delivery of miRNA-30a mimic via cyclo(RGDfC)-gated polymeric-nanoplexes with dendrimer templates to alleviate podocyte conditions. The nanoplexes able to protect RNase enzyme and to exhibit greater cellular uptake viaαvβ3 receptor selective binding in HG treated podocytes. The nanoplexes up-regulated the expression level of miRNA-30a and repress the elevated Notch-1 signaling in HG exposed podocytes. The critical results of in vivo experimentation attribute marked suppression of Notch-1 in streptozotocin (STZ) induced diabetic C57BL/6 mice and reduced glomerular expansion and fibrosis in the glomerular area. Developed nanoplexes represents an efficient platform for the targeted delivery of exogenous miRNA to podocytes. The approach developed herein could be extrapolated to other gene therapeutics and other kidney-related diseases.
Collapse
|
47
|
Cell-Penetrating Peptides and Transportan. Pharmaceutics 2021; 13:pharmaceutics13070987. [PMID: 34210007 PMCID: PMC8308968 DOI: 10.3390/pharmaceutics13070987] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/21/2022] Open
Abstract
In the most recent 25–30 years, multiple novel mechanisms and applications of cell-penetrating peptides (CPP) have been demonstrated, leading to novel drug delivery systems. In this review, I present a brief introduction to the CPP area with selected recent achievements. This is followed by a nostalgic journey into the research in my own laboratories, which lead to multiple CPPs, starting from transportan and paving a way to CPP-based therapeutic developments in the delivery of bio-functional materials, such as peptides, proteins, vaccines, oligonucleotides and small molecules, etc.
Collapse
|
48
|
Wang J, Wang X, Gao Y, Lin Z, Chen J, Gigantelli J, Shapiro JI, Xie Z, Pierre SV. Stress Signal Regulation by Na/K-ATPase As a New Approach to Promote Physiological Revascularization in a Mouse Model of Ischemic Retinopathy. Invest Ophthalmol Vis Sci 2021; 61:9. [PMID: 33275652 PMCID: PMC7718810 DOI: 10.1167/iovs.61.14.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose The identification of target pathways to block excessive angiogenesis while simultaneously restoring physiological vasculature is an unmet goal in the therapeutic management of ischemic retinopathies. pNaKtide, a cell-permeable peptide that we have designed by mapping the site of α1 Na/K-ATPase (NKA)/Src binding, blocks the formation of α1 NKA/Src/reactive oxygen species (ROS) amplification loops and restores physiological ROS signaling in a number of oxidative disease models. The aim of this study was to evaluate the importance of the NKA/Src/ROS amplification loop and the effect of pNaKtide in experimental ischemic retinopathy. Methods Human retinal microvascular endothelial cells (HRMECs) and retinal pigment epithelium (ARPE-19) cells were used to evaluate the effect of pNaKtide on viability, proliferation, and angiogenesis. Retinal toxicity and distribution were assessed in those cells and in the mouse. Subsequently, the role and molecular mechanism of NKA/Src in ROS stress signaling were evaluated biochemically in the retinas of mice exposed to the well-established protocol of oxygen-induced retinopathy (OIR). Finally, pNaKtide efficacy was assessed in this model. Results The results suggest a key role of α1 NKA in the regulation of ROS stress and the Nrf2 pathway in mouse OIR retinas. Inhibition of α1 NKA/Src by pNaKtide reduced pathologic ROS signaling and restored normal expression of hypoxia-inducible factor 1-α/vascular endothelial growth factor (VEGF). Unlike anti-VEGF agents, pNaKtide did promote retinal revascularization while inhibiting neovascularization and inflammation. Conclusions Targeting α1 NKA represents a novel strategy to develop therapeutics that not only inhibit neovascularization but also promote physiological revascularization in ischemic eye diseases.
Collapse
Affiliation(s)
- Jiayan Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Xiaoliang Wang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States.,Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Yingnyu Gao
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Zhucheng Lin
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - James Gigantelli
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Joseph I Shapiro
- Departments of Medicine, Ophthalmology, Pharmacology, and Surgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| |
Collapse
|
49
|
Nomura TK, Heishima K, Sugito N, Sugawara R, Ueda H, Yukihiro A, Honda R. Specific inhibition of oncogenic RAS using cell-permeable RAS-binding domains. Cell Chem Biol 2021; 28:1581-1589.e6. [PMID: 33964212 DOI: 10.1016/j.chembiol.2021.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/17/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Oncogenic RAS proteins, common oncogenic drivers in many human cancers, have been refractory to conventional small-molecule and macromolecule inhibitors due to their intracellular localization and the lack of druggable pockets. Here, we present a feasible strategy for designing RAS inhibitors that involves intracellular delivery of RAS-binding domain (RBD), a nanomolar-affinity specific ligand of RAS. Screening of 51 different combinations of RBD and cell-permeable peptides has identified Pen-cRaf-v1 as a cell-permeable pan-RAS inhibitor capable of targeting both G12C and non-G12C RAS mutants. Pen-cRaf-v1 crosses the cell membrane via endocytosis, competitively inhibits RAS-effector interaction, and thereby exerts anticancer activity against several KRAS-mutant cancer cell lines. Moreover, Pen-cRaf-v1 exhibits excellent activity comparable with a leading pan-RAS inhibitor (BI-2852), as well as high target specificity in transcriptome analysis and alanine mutation analysis. These findings demonstrate that specific inhibition of oncogenic RAS, and possibly treatment of RAS-mutant cancer, is feasible by intracellular delivery of RBD.
Collapse
Affiliation(s)
- Teiko Komori Nomura
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Kazuki Heishima
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Nobuhiko Sugito
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Ryota Sugawara
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Hiroshi Ueda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Akao Yukihiro
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan
| | - Ryo Honda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1193, Japan.
| |
Collapse
|
50
|
Walker DR, Alizadehmojarad AA, Kolomeisky AB, Hartgerink JD. Charge-Free, Stabilizing Amide-π Interactions Can Be Used to Control Collagen Triple-Helix Self-Assembly. Biomacromolecules 2021; 22:2137-2147. [PMID: 33881314 DOI: 10.1021/acs.biomac.1c00234] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There is a noted lack of understood, controllable interactions for directing the organization of collagen triple helices. While the field has had success using charge-pair interactions and cation-π interactions in helix design, these alone are not adequate for achieving the degree of specificity desirable for these supramolecular structures. Furthermore, because of the reliance on electrostatic interactions, designed heterotrimeric systems have been heavily charged, a property undesirable in some applications. Amide-π interactions are a comparatively understudied class of charge-free interactions, which could potentially be harnessed for triple-helix design. Herein, we propose, validate, and utilize pairwise amino acid amide-π interactions in collagen triple-helix design. Glutamine-phenylalanine pairs, when arranged in an axial geometry, are found to exhibit a moderately stabilizing effect, while in the lateral geometry, this pair is destabilizing. Together this allows glutamine-phenylalanine pairs to effectively set the register of triple helices. In contrast, interactions between asparagine and phenylalanine appear to have little effect on triple-helical stability. After deconvoluting the contributions of these amino acids to triple-helix stability, we demonstrate these new glutamine-phenylalanine interactions in the successful design of a heterotrimeric triple helix. The results of all of these analyses are used to update our collagen triple-helix thermal stability prediction algorithm, Scoring function for Collagen Emulating Peptides' Temperature of Transition (SCEPTTr).
Collapse
|