1
|
Ghosh P, Seitz O. Boronic Acid-Linked Apo-Zinc Finger Protein for Ubiquitin Delivery in Live Cells. Chembiochem 2025; 26:e202401040. [PMID: 39950407 DOI: 10.1002/cbic.202401040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
Delivering cargo into live cells has extensive applications in chemistry, biology, and medicine. Cell-penetrating peptides (CPPs) provide an ideal solution for cellular delivery. Enhancing CPPs with additional functional units can improve delivery efficiency. We investigate the conjugation of boronic acid modules to enhance internalization through interactions with cell surface glycans. The aim of this study is to determine whether adding boronic acid can transform a peptide that typically lacks CPP properties into one that functions as a CPP, enabling the delivery of crucial biological cargo like ubiquitin (Ub). The zinc finger protein in its apo state was selected as a "boronate-enabled" CPP. Results indicate that skeletal point mutations and post-synthetic modifications, combined with conjugated benzoboroxole derivatives, enable the apo-ZFP the ability to transport Ub within A549 cells, confirmed through microscopy and flow cytometry. This effective internalization of cargo offers valuable insights for advancing the development of boronic acid-mediated cell-penetrating peptides.
Collapse
Affiliation(s)
- Pritam Ghosh
- Institute of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
| | - Oliver Seitz
- Institute of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
| |
Collapse
|
2
|
Sun Y, Wu X, Li J, Radiom M, Mezzenga R, Verma CS, Yu J, Miserez A. Phase-separating peptide coacervates with programmable material properties for universal intracellular delivery of macromolecules. Nat Commun 2024; 15:10094. [PMID: 39572548 PMCID: PMC11582321 DOI: 10.1038/s41467-024-54463-z] [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: 07/05/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Phase-separating peptides (PSPs) self-assembling into coacervate microdroplets (CMs) are a promising class of intracellular delivery vehicles that can release macromolecular modalities deployed in a wide range of therapeutic treatments. However, the molecular grammar governing intracellular uptake and release kinetics of CMs remains elusive. Here, we systematically manipulate the sequence of PSPs to unravel the relationships between their molecular structure, the physical properties of the resulting CMs, and their delivery efficacy. We show that a few amino acid alterations are sufficient to modulate the viscoelastic properties of CMs towards either a gel-like or a liquid-like state as well as their binding interaction with cellular membranes, collectively enabling to tune the kinetics of intracellular cargo release. We also demonstrate that the optimized PSPs CMs display excellent transfection efficiency in hard-to-transfect cells such as primary fibroblasts and immune cells. Our findings provide molecular guidelines to precisely program the material properties of PSP CMs and achieve tunable cellular uptake and release kinetics depending on the cargo modality, with broad implications for therapeutic applications such as protein, gene, and immune cell therapies.
Collapse
Affiliation(s)
- Yue Sun
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
| | - Xi Wu
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
| | - Jianguo Li
- Bioinformatics Institute, Agency for Science, Technology and Research, 30 Biopolis Street, Matrix, 138671, Singapore, Singapore
- Singapore Eye Research Institute, 169856, Singapore, Singapore
| | - Milad Radiom
- Department of Health Sciences & Technology, ETH Zurich, 8092, Zürich, Switzerland
| | - Raffaele Mezzenga
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
- Department of Health Sciences & Technology, ETH Zurich, 8092, Zürich, Switzerland
- Department of Materials, ETH Zurich, 8092, Zürich, Switzerland
| | - Chandra Shekhar Verma
- Bioinformatics Institute, Agency for Science, Technology and Research, 30 Biopolis Street, Matrix, 138671, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, 117558, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore, Singapore
| | - Jing Yu
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore, Singapore
| | - Ali Miserez
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, 639798, Singapore, Singapore.
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore, Singapore.
| |
Collapse
|
3
|
Dowaidar M. Uptake pathways of cell-penetrating peptides in the context of drug delivery, gene therapy, and vaccine development. Cell Signal 2024; 117:111116. [PMID: 38408550 DOI: 10.1016/j.cellsig.2024.111116] [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: 01/03/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
Cell-penetrating peptides have been extensively utilized for the purpose of facilitating the intracellular delivery of cargo that is impermeable to the cell membrane. The researchers have exhibited proficient delivery capabilities for oligonucleotides, thereby establishing cell-penetrating peptides as a potent instrument in the field of gene therapy. Furthermore, they have demonstrated a high level of efficiency in delivering several additional payloads. Cell penetrating peptides (CPPs) possess the capability to efficiently transport therapeutic molecules to specific cells, hence offering potential remedies for many illnesses. Hence, their utilization is imperative for the improvement of therapeutic vaccines. In contemporary studies, a plethora of cell-penetrating peptides have been unveiled, each characterized by its own distinct structural attributes and associated mechanisms. Although it is widely acknowledged that there are multiple pathways through which particles might be internalized, a comprehensive understanding of the specific mechanisms by which these particles enter cells has to be fully elucidated. The absorption of cell-penetrating peptides can occur through either direct translocation or endocytosis. However, it is worth noting that categories of cell-penetrating peptides are not commonly linked to specific entrance mechanisms. Furthermore, research has demonstrated that cell-penetrating peptides (CPPs) possess the capacity to enhance antigen uptake by cells and facilitate the traversal of various biological barriers. The primary objective of this work is to examine the mechanisms by which cell-penetrating peptides are internalized by cells and their significance in facilitating the administration of drugs, particularly in the context of gene therapy and vaccine development. The current study investigates the immunostimulatory properties of numerous vaccine components administered using different cell-penetrating peptides (CPPs). This study encompassed a comprehensive discussion on various topics, including the uptake pathways and mechanisms of cell-penetrating peptides (CPPs), the utilization of CPPs as innovative vectors for gene therapy, the role of CPPs in vaccine development, and the potential of CPPs for antigen delivery in the context of vaccine development.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia.
| |
Collapse
|
4
|
Saha A, Mandal S, Arafiles JVV, Gómez‐González J, Hackenberger CPR, Brik A. Structure-Uptake Relationship Study of DABCYL Derivatives Linked to Cyclic Cell-Penetrating Peptides for Live-Cell Delivery of Synthetic Proteins. Angew Chem Int Ed Engl 2022; 61:e202207551. [PMID: 36004945 PMCID: PMC9828537 DOI: 10.1002/anie.202207551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Indexed: 01/12/2023]
Abstract
Modifying cyclic cell-penetrating deca-arginine (cR10) peptides with 4-(4-dimethylaminophenylazo)benzoic acid (DABCYL) improves the uptake efficiency of synthetic ubiquitin (Ub) cargoes into living cells. To probe the role of the DABCYL moiety, we performed time-lapse microscopy and fluorescence lifetime imaging microscopy (FLIM) of fluorescent DABCYL-R10 to evaluate the impact on cell entry by the formation of nucleation zones. Furthermore, we performed a structure-uptake relationship study with 13 DABCYL derivatives coupled to CPP to examine their effect on the cell-uptake efficiency when conjugated to mono-Ub through disulfide linkages. Our results show that through structure variations of the DABCYL moiety alone we could reach, at nanomolar concentration, an additional threefold increase in the cytosolic delivery of Ub, which will enable studies on various intracellular processes related to Ub signaling.
Collapse
Affiliation(s)
- Abhishek Saha
- Schulich Faculty of ChemistryTechnion-Israel Institute of TechnologyHaifa3200008Israel
| | - Shaswati Mandal
- Schulich Faculty of ChemistryTechnion-Israel Institute of TechnologyHaifa3200008Israel
| | - Jan Vincent V. Arafiles
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)Robert-Rössle-Strasse 10Berlin13125Germany
| | - Jacobo Gómez‐González
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)Robert-Rössle-Strasse 10Berlin13125Germany
| | - Christian P. R. Hackenberger
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)Robert-Rössle-Strasse 10Berlin13125Germany
- Department of ChemistryHumboldt Universität zu BerlinBrook-Taylor-Str.2Berlin12489Germany
| | - Ashraf Brik
- Schulich Faculty of ChemistryTechnion-Israel Institute of TechnologyHaifa3200008Israel
| |
Collapse
|
5
|
Hasannejad-Asl B, Pooresmaeil F, Takamoli S, Dabiri M, Bolhassani A. Cell penetrating peptide: A potent delivery system in vaccine development. Front Pharmacol 2022; 13:1072685. [PMID: 36425579 PMCID: PMC9679422 DOI: 10.3389/fphar.2022.1072685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 07/28/2023] Open
Abstract
One of the main obstacles to most medication administrations (such as the vaccine constructs) is the cellular membrane's inadequate permeability, which reduces their efficiency. Cell-penetrating peptides (CPPs) or protein transduction domains (PTDs) are well-known as potent biological nanocarriers to overcome this natural barrier, and to deliver membrane-impermeable substances into cells. The physicochemical properties of CPPs, the attached cargo, concentration, and cell type substantially influence the internalization mechanism. Although the exact mechanism of cellular uptake and the following processing of CPPs are still uncertain; but however, they can facilitate intracellular transfer through both endocytic and non-endocytic pathways. Improved endosomal escape efficiency, selective cell targeting, and improved uptake, processing, and presentation of antigen by antigen-presenting cells (APCs) have been reported by CPPs. Different in vitro and in vivo investigations using CPP conjugates show their potential as therapeutic agents in various medical areas such as infectious and non-infectious disorders. Effective treatments for a variety of diseases may be provided by vaccines that can cooperatively stimulate T cell-mediated immunity (T helper cell activity or cytotoxic T cell function), and immunologic memory. Delivery of antigen epitopes to APCs, and generation of a potent immune response is essential for an efficacious vaccine that can be facilitated by CPPs. The current review describes the delivery of numerous vaccine components by various CPPs and their immunostimulatory properties.
Collapse
Affiliation(s)
- Behnam Hasannejad-Asl
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti, University of Medical Sciences, Tehran, Iran
| | - Farkhondeh Pooresmaeil
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Shahla Takamoli
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Mehran Dabiri
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Geng J, Xia X, Teng L, Wang L, Chen L, Guo X, Belingon B, Li J, Feng X, Li X, Shang W, Wan Y, Wang H. Emerging landscape of cell-penetrating peptide-mediated nucleic acid delivery and their utility in imaging, gene-editing, and RNA-sequencing. J Control Release 2022; 341:166-183. [PMID: 34822907 DOI: 10.1016/j.jconrel.2021.11.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022]
Abstract
The safety issues like immunogenicity and unacceptable cancer risk of viral vectors for DNA/mRNA vaccine delivery necessitate the development of non-viral vectors with no toxicity. Among the non-viral strategies, cell-penetrating peptides (CPPs) have been a topic of interest recently because of their ability to cross plasma membranes and facilitate nucleic acids delivery both in vivo and in vitro. In addition to the application in the field of gene vaccine and gene therapy, CPPs based nucleic acids delivery have been proved by its potential application like gene editing, RNA-sequencing, and imaging. Here, we focus on summarizing the recent applications and progress of CPPs-mediated nucleic acids delivery and discuss the current problems and solutions in this field.
Collapse
Affiliation(s)
- Jingping Geng
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Xuan Xia
- Department of Physiology and Pathophysiology, Medical School, China Three Gorges University, Yichang 443002, China
| | - Lin Teng
- Department of Cardiovascular Medicine, The First Clinical Medical College of China Three Gorges University, Yichang 443002, China
| | - Lidan Wang
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Linlin Chen
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; Affiliated Ren He Hospital of China Three Gorges University, Yichang 443002, China
| | - Xiangli Guo
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Bonn Belingon
- Institute of Cell Engineering, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Jason Li
- Department of Biology, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Xuemei Feng
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Xianghui Li
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Wendou Shang
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Yingying Wan
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Hu Wang
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
7
|
Ali S, Dussouillez C, Padilla B, Frisch B, Mason AJ, Kichler A. Design of a new cell penetrating peptide for DNA, siRNA and mRNA delivery. J Gene Med 2021; 24:e3401. [PMID: 34856643 DOI: 10.1002/jgm.3401] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Delivery systems, including peptide-based ones, that destabilize endosomes in a pH-dependent manner are increasingly used to deliver cargoes of therapeutic interest, such as nucleic acids and proteins into mammalian cells. METHODS The negatively charged amphipathic alpha-helicoidal forming peptide named HELP (Helical Erythrocyte Lysing Peptide) is a derivative from the bee venom melittin and was shown to have a pH-dependent activity with the highest lytic activity at pH 5.0 at the same time as becoming inactive when the pH is increased. The present study aimed to determine whether replacement in the HELP peptide of the glutamic acid residues by histidines, for which the protonation state is sensitive to the pH changes that occur during endosomal acidification, can transform this fusogenic peptide into a carrier able to deliver different nucleic acids into mammalian cells. RESULTS The resulting HELP-4H peptide displays high plasmid DNA, small interfering RNA and mRNA delivery capabilities. Importantly, in contrast to other cationic peptides, its transfection activity was only marginally affected by the presence of serum. Using circular dichroism, we found that acidic pH did not induce significant conformational changes for HELP-4H. CONCLUSIONS In summary, we were able to develop a new cationic histidine rich peptide able to efficiently deliver various nucleic acids into cells.
Collapse
Affiliation(s)
- Salif Ali
- 3Bio Team, CAMB 7199 CNRS - University of Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Candice Dussouillez
- 3Bio Team, CAMB 7199 CNRS - University of Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Beatriz Padilla
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, London, UK
| | - Benoît Frisch
- 3Bio Team, CAMB 7199 CNRS - University of Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, London, UK
| | - Antoine Kichler
- 3Bio Team, CAMB 7199 CNRS - University of Strasbourg, Faculté de Pharmacie, Illkirch, France
| |
Collapse
|
8
|
Alhakamy NA, Ahmed OAA, Md S, Fahmy UA. Mastoparan, a Peptide Toxin from Wasp Venom Conjugated Fluvastatin Nanocomplex for Suppression of Lung Cancer Cell Growth. Polymers (Basel) 2021; 13:4225. [PMID: 34883728 PMCID: PMC8659920 DOI: 10.3390/polym13234225] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Lung cancer has a very low survival rate, and non-small cell lung cancer comprises around 85% of all types of lung cancers. Fluvastatin (FLV) has demonstrated the apoptosis and suppression of tumor-cell proliferation against lung cancer cells in vitro. Drug-peptide nanoconjugates were found to enhance the cytotoxicity of anti-cancer drugs. Thus, the present study aimed to develop a nanocomplex of FLV with mastoparan (MAS), which is a peptide that has membranolytic anti-tumor activity. The nanocomplex of FLV and MAS (MAS-FLV-NC) was prepared and optimized for particle size using Box-Behnken design. The amount of FLV had the highest influence on particle size. While higher levels of FLV and incubation time favored higher particle size, a higher level of sonication time reduced the particle size of MAS-FLV-NC. The optimum formula of MAS-FLV-NC used 1.00 mg of FLV and was prepared with an incubation time of 12.1339 min and a sonication time of 6 min. The resultant particle size was 77.648 nm. The in vitro cell line studies of MAS-FLV-NC, FLV, and MAS were carried out in A549 cells. The IC50 values of MAS-FLV-NC, FLV, and MAS were 18.6 ± 0.9, 58.4 ± 2.8, and 34.3 ± 1.6 µg/mL respectively, showing the enhanced cytotoxicity of MAS-FLV-NC. The apoptotic activity showed that MAS-FLV-NC produced a higher percentage of cells in the late phase, showing a higher apoptotic activity than FLV and MAS. Furthermore, cell cycle arrest in S and Pre G1 phases by MAS-FLV-NC was observed in the cell cycle analysis by flow cytometry. The loss of mitochondrial membrane potential after MAS-FLV-NC treatment was significantly higher than those observed for FLV and MAS. The IL-1β, IL-6, and NF-kB expressions were inhibited, whereas TNF-α, caspase-3, and ROS expressions were enhanced by MAS-FLV-NC treatment. Furthermore, the expression levels of Bax, Bcl-2, and p53 strongly established the enhanced cytotoxic effect of MAS-FLV-NC. The results indicated that MAS-FLV-NC has better cytotoxicity than individual effects of MAS and FLV in A549 cells. Further pre-clinical and clinical studies are needed for developing MAS-FLV-NC to a clinically successful therapeutic approach against lung cancer.
Collapse
Affiliation(s)
- Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (S.M.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Osama A. A. Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (S.M.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (S.M.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Usama A. Fahmy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (O.A.A.A.); (S.M.)
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
9
|
Kardani K, Milani A, H Shabani S, Bolhassani A. Cell penetrating peptides: the potent multi-cargo intracellular carriers. Expert Opin Drug Deliv 2019; 16:1227-1258. [PMID: 31583914 DOI: 10.1080/17425247.2019.1676720] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Cell penetrating peptides (CPPs) known as protein translocation domains (PTD), membrane translocating sequences (MTS), or Trojan peptides (TP) are able to cross biological membranes without clear toxicity using different mechanisms, and facilitate the intracellular delivery of a variety of bioactive cargos. CPPs could overcome some limitations of drug delivery and combat resistant strains against a broad range of diseases. Despite delivery of different therapeutic molecules by CPPs, they lack cell specificity and have a short duration of action. These limitations led to design of combined cargo delivery systems and subsequently improvement of their clinical applications. Areas covered: This review covers all our studies and other researchers in different aspects of CPPs such as classification, uptake mechanisms, and biomedical applications. Expert opinion: Due to low cytotoxicity of CPPs as compared to other carriers and final degradation to amino acids, they are suitable for preclinical and clinical studies. Generally, the efficiency of CPPs was suitable to penetrate the cell membrane and deliver different cargos to specific intracellular sites. However, no CPP-based therapeutic approach has approved by FDA, yet; because there are some disadvantages for CPPs including short half-life in blood, and nonspecific CPP-mediated delivery to normal tissue. Thus, some methods were used to develop the functions of CPPs in vitro and in vivo including the augmentation of cell specificity by activatable CPPs, specific transport into cell organelles by insertion of corresponding localization sequences, incorporation of CPPs into multifunctional dendrimeric or liposomal nanocarriers to improve selectivity and efficiency especially in tumor cells.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Samaneh H Shabani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| |
Collapse
|
10
|
Rostami B, Irani S, Bolhassani A, Cohan RA. M918: A Novel Cell Penetrating Peptide for Effective Delivery of HIV-1 Nef and Hsp20-Nef Proteins into Eukaryotic Cell Lines. Curr HIV Res 2019; 16:280-287. [PMID: 30520377 PMCID: PMC6416460 DOI: 10.2174/1570162x17666181206111859] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/30/2018] [Accepted: 12/02/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND HIV-1 Nef protein is a possible attractive target in the development of therapeutic HIV vaccines including protein-based vaccines. The most important disadvantage of protein-based vaccines is their low immunogenicity which can be improved by heat shock proteins (Hsps) as an immunomodulator, and cell-penetrating peptides (CPPs) as a carrier. METHODS In this study, the HIV-1 Nef and Hsp20-Nef proteins were generated in E.coli expression system for delivery into the HEK-293T mammalian cell line using a novel cell-penetrating peptide, M918, in a non-covalent fashion. The size, zeta potential and morphology of the peptide/protein complexes were studied by scanning electron microscopy (SEM) and Zeta sizer. The efficiency of Nef and Hsp20-Nef transfection using M918 was evaluated by western blotting in HEK-293T cell line. RESULTS The SEM data confirmed the formation of discrete nanoparticles with a diameter of approximately 200-250 nm and 50-80 nm for M918/Nef and M918/Hsp20-Nef, respectively. The dominant band of ~ 27 kDa and ~ 47 kDa was detected in the transfected cells with the Nef/ M918 and Hsp20-Nef/ M918 nanoparticles at a molar ratio of 1:20 using anti-HIV-1 Nef monoclonal antibody. These bands were not detected in the un-transfected and transfected cells with Nef or Hsp20- Nef protein alone indicating that M918 could increase the penetration of Nef and Hsp20-Nef proteins into the cells. CONCLUSION These data suggest that M918 CPP can be used to enter HIV-1 Nef and Hsp20-Nef proteins inside mammalian cells efficiently as a promising approach in HIV-1 vaccine development.
Collapse
Affiliation(s)
- Bahareh Rostami
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Pilot Nano-Biotechnology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
Schober T, Wehl I, Afonin S, Babii O, Iampolska A, Schepers U, Komarov IV, Ulrich AS. Controlling the Uptake of Diarylethene‐Based Cell‐Penetrating Peptides into Cells Using Light. CHEMPHOTOCHEM 2019. [DOI: 10.1002/cptc.201900019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tim Schober
- Karlsruhe Institute of Technology (KIT)Institute of Organic Chemistry (IOC) Fritz-Haber-Weg 6 76131 Karlsruhe Germany
| | - Ilona Wehl
- KIT, Institute of Functional Interfaces (IFG) POB 3640 76021 Karlsruhe Germany
| | - Sergii Afonin
- KIT, Institute of Biological Interfaces (IBG-2) POB 3640 76021 Karlsruhe Germany
| | - Oleg Babii
- KIT, Institute of Biological Interfaces (IBG-2) POB 3640 76021 Karlsruhe Germany
| | - Anna Iampolska
- Taras Shevchenko National University of Kyiv Vul. Volodymyrska 60 01601 Kyiv Ukraine
- Enamine Ltd. Vul. Chervonotkatska 78 02094 Kyiv Ukraine
| | - Ute Schepers
- KIT, Institute of Functional Interfaces (IFG) POB 3640 76021 Karlsruhe Germany
| | - Igor V. Komarov
- Taras Shevchenko National University of Kyiv Vul. Volodymyrska 60 01601 Kyiv Ukraine
- Lumobiotics GmbH Auerstraße 2 76227 Karlsruhe Germany
| | - Anne S. Ulrich
- Karlsruhe Institute of Technology (KIT)Institute of Organic Chemistry (IOC) Fritz-Haber-Weg 6 76131 Karlsruhe Germany
- KIT, Institute of Biological Interfaces (IBG-2) POB 3640 76021 Karlsruhe Germany
| |
Collapse
|
12
|
Donahue ND, Acar H, Wilhelm S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv Drug Deliv Rev 2019; 143:68-96. [PMID: 31022434 DOI: 10.1016/j.addr.2019.04.008] [Citation(s) in RCA: 572] [Impact Index Per Article: 95.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/14/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
Abstract
Nanoparticle-based therapeutics and diagnostics are commonly referred to as nanomedicine and may significantly impact the future of healthcare. However, the clinical translation of these technologies is challenging. One of these challenges is the efficient delivery of nanoparticles to specific cell populations and subcellular targets in the body to elicit desired biological and therapeutic responses. It is critical for researchers to understand the fundamental concepts of how nanoparticles interact with biological systems to predict and control in vivo nanoparticle transport for improved clinical benefit. In this overview article, we review and discuss cellular internalization pathways, summarize the field`s understanding of how nanoparticle physicochemical properties affect cellular interactions, and explore and discuss intracellular nanoparticle trafficking and kinetics. Our overview may provide a valuable resource for researchers and may inspire new studies to expand our current understanding of nanotechnology-biology interactions at cellular and subcellular levels with the goal to improve clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Nathan D Donahue
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Handan Acar
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States; Stephenson Cancer Center, Oklahoma City, Oklahoma 73104, United States.
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States; Stephenson Cancer Center, Oklahoma City, Oklahoma 73104, United States.
| |
Collapse
|
13
|
Sönmez M, Ficai D, Ficai A, Alexandrescu L, Georgescu M, Trusca R, Gurau D, Titu MA, Andronescu E. Applications of mesoporous silica in biosensing and controlled release of insulin. Int J Pharm 2018; 549:179-200. [PMID: 30016674 DOI: 10.1016/j.ijpharm.2018.07.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 01/22/2023]
Abstract
The development of new oral insulin delivery systems could bring significant benefits to insulin-dependent patients due to the simplicity of the method, avoidance of pain caused by parenteral administration and maintenance of optimal therapeutic levels for a longer period. However, administration of such therapeutic proteins orally remains a challenge because insulin (Ins) is a very sensitive molecule and can be easily degraded under the existing pH conditions in the stomach and intestines. Moreover, due to the large size of insulin, intestinal epithelium permeability is very low. This could be improved by immobilizing insulin in the mesoporous silica pores (MSN), acting as a shield to protect the molecule integrity from the proteolytic degradation existing in the stomach and upper part of the small intestine. Due to the high adsorption capacity of insulin, biocompatibility, ease of functionalization with various organic and/or inorganic groups, high mechanical and chemical resistance, adjustable pore size and volume, MSN is considered an ideal candidate for the development of controlled release systems that are sensitive to various stimuli (pH, temperature) as well as to glucose. Modifying MSN surfaces by coating with various mucoadhesive polymers (chitosan, alginate, etc.) will also facilitate interaction with the intestinal mucus and improve intestinal retention time. Moreover, the development of glucose-responsive systems for achieving MSN-based self-regulated insulin delivery, decorated with various components serving as sensors - glucose oxidase (GODx) and phenylboronic acid (PBA) that can control the insulin dosage, avoiding overdose leading to serious hypoglycemia. MSN have also been tested for application as biosensors for glucose monitoring.
Collapse
Affiliation(s)
- Maria Sönmez
- Research Institute of the University of Bucharest, 36-46 bd. M. Kogalniceanu, Bucharest, Romania
| | - Denisa Ficai
- Politehnica University of Bucharest, Faculty of Applied Chemistry and Material Science, 1-7 Polizu St., Bucharest, Romania
| | - Anton Ficai
- S.C. Metav R&D S.A, 31 C.A. Rosetti Str., Bucharest, Romania
| | - Laurentia Alexandrescu
- National Research & Development Institute for Textiles and Leather-Division: Leather and Footwear Research Institute, 93 Ion Minulescu St., Bucharest, Romania
| | - Mihai Georgescu
- National Research & Development Institute for Textiles and Leather-Division: Leather and Footwear Research Institute, 93 Ion Minulescu St., Bucharest, Romania
| | - Roxana Trusca
- S.C. Metav R&D S.A, 31 C.A. Rosetti Str., Bucharest, Romania
| | - Dana Gurau
- National Research & Development Institute for Textiles and Leather-Division: Leather and Footwear Research Institute, 93 Ion Minulescu St., Bucharest, Romania
| | | | - Ecaterina Andronescu
- Politehnica University of Bucharest, Faculty of Applied Chemistry and Material Science, 1-7 Polizu St., Bucharest, Romania.
| |
Collapse
|
14
|
Gatta AK, Hariharapura RC, Udupa N, Reddy MS, Josyula VR. Strategies for improving the specificity of siRNAs for enhanced therapeutic potential. Expert Opin Drug Discov 2018; 13:709-725. [PMID: 29902093 DOI: 10.1080/17460441.2018.1480607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION RNA interference has become a tool of choice in the development of drugs in various therapeutic areas of Post Transcriptional Gene Silencing (PTGS). The critical element in developing successful RNAi therapeutics lies in designing small interfering RNA (siRNA) using an efficient algorithm satisfying the designing criteria. Further, translation of siRNA from bench-side to bedside needs an efficient delivery system and/or chemical modification. Areas covered: This review emphasizes the importance of dicer, the criteria for efficient siRNA design, the currently available algorithms and strategies to overcome off-target effects, immune stimulatory effects and endosomal trap. Expert opinion: Specificity and stability are the primary concerns for siRNA therapeutics. The design criteria and algorithms should be chosen rationally to have a siRNA sequence that binds to the corresponding mRNA as it happens in the Watson and Crick base pairing. However, it must evade a few more hurdles (Endocytosis, Serum stability etc.) to be functional in the cytosol.
Collapse
Affiliation(s)
- Aditya Kiran Gatta
- a Cell and Molecular Biology lab, Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Raghu Chandrashekhar Hariharapura
- a Cell and Molecular Biology lab, Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Nayanabhirama Udupa
- b Research Directorate of Health Sciences , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Meka Sreenivasa Reddy
- c Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Venkata Rao Josyula
- a Cell and Molecular Biology lab, Department of Pharmaceutical Biotechnology , Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal , Karnataka , India
| |
Collapse
|
15
|
Barattin M, Mattarei A, Balasso A, Paradisi C, Cantù L, Del Favero E, Viitala T, Mastrotto F, Caliceti P, Salmaso S. pH-Controlled Liposomes for Enhanced Cell Penetration in Tumor Environment. ACS APPLIED MATERIALS & INTERFACES 2018; 10:17646-17661. [PMID: 29737834 DOI: 10.1021/acsami.8b03469] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
An innovative pH-switchable colloidal system that can be exploited for site-selective anticancer drug delivery has been generated by liposome decoration with a new novel synthetic non-peptidic oligo-arginine cell-penetration enhancer (CPE) and a quenching PEGylated counterpart that detaches from the vesicle surface under the acidic conditions of tumors. The CPE module ( Arg4- DAG) is formed by four arginine units conjugated to a first-generation (G1) 2,2-bis(hydroxymethyl)propionic acid (bis-MPA)/2,2-bis(aminomethyl)propionic acid (bis-AMPA) polyester dendron terminating with 1,2-distearoyl-3-azidopropane for liposome bilayer insertion. The zeta potential of the Arg4- DAG-decorated liposomes increased up to +32 mV as the Arg4- DAG/lipids molar ratio increased. The Arg4- DAG liposome shielding at pH 7.4 was provided by methoxy-PEG5 kDa-polymethacryloyl sulfadimethoxine (mPEG5 kDa-SDM8) with 7.1 apparent p Ka. Zeta potential, surface plasmon resonance and synchrotron small-angle X-ray scattering analyses showed that at pH 7.4 mPEG5 kDa-SDM8 associates with polycationic Arg4- DAG-decorated liposomes yielding liposomes with neutral zeta potential. At pH 6.5, which mimics the tumor environment, mPEG5 kDa-SDM8 detaches from the liposome surface yielding Arg4- DAG exposure. Flow cytometry and confocal microscopy showed a 30-fold higher HeLa cancer cell association of the Arg4- DAG-decorated liposomes compared to non-decorated liposomes. At pH 7.4, the mPEG5 kDa-SDM8-coated liposomes undergo low cell association while remarkable cell association occurred at pH 6.5, which allowed for the controlled intracellular delivery of model macromolecules and small molecules loaded in the liposome under tumor conditions.
Collapse
Affiliation(s)
- Michela Barattin
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Andrea Mattarei
- Department of Chemical Sciences , University of Padova , Via F. Marzolo 1 , Padova 35131 , Italy
| | - Anna Balasso
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Cristina Paradisi
- Department of Chemical Sciences , University of Padova , Via F. Marzolo 1 , Padova 35131 , Italy
| | - Laura Cantù
- Department of Medical Biotechnologies and Traslational Medicine , University of Milano , LITA, Via F.lli Cervi, 93 , Segrate 20090 , Italy
| | - Elena Del Favero
- Department of Medical Biotechnologies and Traslational Medicine , University of Milano , LITA, Via F.lli Cervi, 93 , Segrate 20090 , Italy
| | - Tapani Viitala
- Centre for Drug Research and Division of Pharmaceutical Biosciences, Faculty of Pharmacy , University of Helsinki , Viikinkaari 5 , Helsinki FI-00014 , Finland
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| |
Collapse
|
16
|
Löwik DWPM. Dodging Endosomes: Effective Cytosolic Antibody Delivery. Chembiochem 2017; 18:2196-2198. [PMID: 28940648 DOI: 10.1002/cbic.201700510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Indexed: 11/11/2022]
Abstract
On the inside: New methodologies for delivering antibodies right into the cytosol of cells either directly across the plasma membrane or by allowing the antibody to escape from endosomes have been proposed recently by the Cardoso/Hackenberger and Futaki groups, respectively.
Collapse
Affiliation(s)
- Dennis W P M Löwik
- Radboud University Nijmegen, Institute for Molecules and Materials, Bio-organic Chemistry, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Wu JL, He XY, Liu BY, Gong MQ, Zhuo RX, Cheng SX. Fusion peptide functionalized hybrid nanoparticles for synergistic drug delivery to reverse cancer drug resistance. J Mater Chem B 2017; 5:4697-4704. [PMID: 32264312 DOI: 10.1039/c7tb00655a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A facile self-assembly strategy was developed to decorate polymer/inorganic hybrid nano-sized drug delivery systems with functional peptides. To enhance drug delivery efficacy and overcome tumor drug resistance, a functional fusion peptide containing an RGD sequence for tumor targeting and an R8 sequence for cell penetration was introduced onto the surface of biotinylated carboxymethyl chitosan/CaCO3 (BCMC/CaCO3) hybrid nanoparticles through biotin-avidin interaction to obtain peptide functionalized nanoparticles (PNP). The peptide functionalization results in improved delivery efficiency and effective inhibition for drug resistant tumor cells. Co-delivery of an anti-cancerous drug (doxorubicin hydrochloride, DOX) and a cyclooxygenase-2 inhibitor (celecoxib, CXB) by PNP can further improve the therapeutic efficiency by effectively down-regulating P-gp expression to reduce P-gp mediated drug efflux and increase intracellular drug accumulation.
Collapse
Affiliation(s)
- Jin-Long Wu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
18
|
Takechi-Haraya Y, Aki K, Tohyama Y, Harano Y, Kawakami T, Saito H, Okamura E. Glycosaminoglycan Binding and Non-Endocytic Membrane Translocation of Cell-Permeable Octaarginine Monitored by Real-Time In-Cell NMR Spectroscopy. Pharmaceuticals (Basel) 2017; 10:ph10020042. [PMID: 28420127 PMCID: PMC5490399 DOI: 10.3390/ph10020042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/27/2017] [Accepted: 04/12/2017] [Indexed: 12/13/2022] Open
Abstract
Glycosaminoglycans (GAGs), which are covalently-linked membrane proteins at the cell surface have recently been suggested to involve in not only endocytic cellular uptake but also non-endocytic direct cell membrane translocation of arginine-rich cell-penetrating peptides (CPPs). However, in-situ comprehensive observation and the quantitative analysis of the direct membrane translocation processes are challenging, and the mechanism therefore remains still unresolved. In this work, real-time in-cell NMR spectroscopy was applied to investigate the direct membrane translocation of octaarginine (R8) into living cells. By introducing 4-trifluoromethyl-l-phenylalanine to the N terminus of R8, the non-endocytic membrane translocation of 19F-labeled R8 (19F-R8) into a human myeloid leukemia cell line was observed at 4 °C with a time resolution in the order of minutes. 19F NMR successfully detected real-time R8 translocation: the binding to anionic GAGs at the cell surface, followed by the penetration into the cell membrane, and the entry into cytosol across the membrane. The NMR concentration analysis enabled quantification of how much of R8 was staying in the respective translocation processes with time in situ. Taken together, our in-cell NMR results provide the physicochemical rationale for spontaneous penetration of CPPs in cell membranes.
Collapse
Affiliation(s)
- Yuki Takechi-Haraya
- Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Japan.
| | - Kenzo Aki
- Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Japan.
| | - Yumi Tohyama
- Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Japan.
| | - Yuichi Harano
- Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Japan.
| | - Toru Kawakami
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Hiroyuki Saito
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Emiko Okamura
- Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Japan.
| |
Collapse
|
19
|
Silva JC, Neto LM, Neves RC, Gonçalves JC, Trentini MM, Mucury-Filho R, Smidt KS, Fensterseifer IC, Silva ON, Lima LD, Clissa PB, Vilela N, Guilhelmelli F, Silva LP, Rangel M, Kipnis A, Silva-Pereira I, Franco OL, Junqueira-Kipnis AP, Bocca AL, Mortari MR. Evaluation of the antimicrobial activity of the mastoparan Polybia-MPII isolated from venom of the social wasp Pseudopolybia vespiceps testacea (Vespidae, Hymenoptera). Int J Antimicrob Agents 2017; 49:167-175. [PMID: 28108242 DOI: 10.1016/j.ijantimicag.2016.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/05/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
Mastoparans, a class of peptides found in wasp venom, have significant effects following a sting as well as useful applications in clinical practice. Among these is their potential use in the control of micro-organisms that cause infectious diseases with a significant impact on society. Thus, the present study describes the isolation and identification of a mastoparan peptide from the venom of the social wasp Pseudopolybia vespiceps and evaluated its antimicrobial profile against bacteria (Staphylococcus aureus and Mycobacterium abscessus subsp. massiliense), fungi (Candida albicans and Cryptococcus neoformans) and in vivo S. aureus infection. The membrane pore-forming ability was also assessed. The mastoparan reduced in vitro and ex vivo mycobacterial growth by 80% at 12.5 µM in infected peritoneal macrophages but did not affect the shape of bacterial cells at the dose tested (6.25 µM). The peptide also showed potent action against S. aureus in vitro (EC50 and EC90 values of 1.83 µM and 2.90 µM, respectively) and reduced the in vivo bacterial load after 6 days of topical treatment (5 mg/kg). Antifungal activity was significant, with EC50 and EC90 values of 12.9 µM and 15.3 µM, respectively, for C. albicans, and 11 µM and 22.70 µM, respectively, for C. neoformans. Peptides are currently attracting interest for their potential in the design of antimicrobial drugs, particularly due to the difficulty of micro-organisms in developing resistance to them. In this respect, Polybia-MPII proved to be highly effective, with a lower haemolysis rate compared with peptides of the same family.
Collapse
Affiliation(s)
- Juliana C Silva
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Lázaro M Neto
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Rogério C Neves
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Jaqueline C Gonçalves
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Monalisa M Trentini
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Ricardo Mucury-Filho
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Karina S Smidt
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Isabel C Fensterseifer
- Center for Proteomics and Biochemical Analyses, Genomic Science and Biotechnology Graduate Program, Catholic University of Brasília, Brasília, Brazil; Molecular Pathology Graduate Program, University of Brasilia, Brasília, Brazil
| | - Osmar N Silva
- Center for Proteomics and Biochemical Analyses, Genomic Science and Biotechnology Graduate Program, Catholic University of Brasília, Brasília, Brazil; Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Lilian D Lima
- Laboratory of Immunopathology, Butantan Institute, Sao Paulo, SP, Brazil
| | - Patricia B Clissa
- Laboratory of Immunopathology, Butantan Institute, Sao Paulo, SP, Brazil
| | - Nathália Vilela
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Fernanda Guilhelmelli
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Luciano P Silva
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil; Laboratory of Mass Spectrometry, Embrapa Genetic Resources and Biotechnology, Brasília, Brazil
| | - Marisa Rangel
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil; Laboratory of Immunopathology, Butantan Institute, Sao Paulo, SP, Brazil
| | - André Kipnis
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Ildinete Silva-Pereira
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Octavio L Franco
- Center for Proteomics and Biochemical Analyses, Genomic Science and Biotechnology Graduate Program, Catholic University of Brasília, Brasília, Brazil; Molecular Pathology Graduate Program, University of Brasilia, Brasília, Brazil; Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil; S-Inova Biotech, Graduate Biotechnology Program, Dom Bosco Catholic University, Campo Grande, Brazil
| | - Ana P Junqueira-Kipnis
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Diseases and Public Heath, Federal University of Goiás, Goiânia, Brazil
| | - Anamelia L Bocca
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Márcia R Mortari
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.
| |
Collapse
|
20
|
Bolhassani A, Jafarzade BS, Mardani G. In vitro and in vivo delivery of therapeutic proteins using cell penetrating peptides. Peptides 2017; 87:50-63. [PMID: 27887988 DOI: 10.1016/j.peptides.2016.11.011] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
The failure of proteins to penetrate mammalian cells or target tumor cells restricts their value as therapeutic tools in a variety of diseases such as cancers. Recently, protein transduction domains (PTDs) or cell penetrating peptides (CPPs) have been shown to promote the delivery of therapeutic proteins or peptides into live cells. The successful delivery of proteins mainly depends on their physicochemical properties. Although, linear cell penetrating peptides are one of the most effective delivery vehicles; but currently, cyclic CPPs has been developed to potently transport bioactive full-length proteins into cells. Up to now, several small protein transduction domains from viral proteins including Tat or VP22 could be fused to other peptides or proteins to entry them in various cell types at a dose-dependent approach. A major disadvantage of PTD-fusion proteins is primary uptake into endosomal vesicles leading to inefficient release of the fusion proteins into the cytosol. Recently, non-covalent complex formation (Chariot) between proteins and CPPs has attracted a special interest to overcome some delivery limitations (e.g., toxicity). Many preclinical and clinical trials of CPP-based delivery are currently under evaluation. Generally, development of more efficient protein transduction domains would significantly increase the potency of protein therapeutics. Moreover, the synergistic or combined effects of CPPs with other delivery systems for protein/peptide drug delivery would promote their therapeutic effects in cancer and other diseases. In this review, we will describe the functions and implications of CPPs for delivering the therapeutic proteins or peptides in preclinical and clinical studies.
Collapse
Affiliation(s)
- Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | | | - Golnaz Mardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
21
|
Farkhani SM, Shirani A, Mohammadi S, Zakeri-Milani P, Shahbazi Mojarrad J, Valizadeh H. Effect of poly-glutamate on uptake efficiency and cytotoxicity of cell penetrating peptides. IET Nanobiotechnol 2016; 10:87-95. [PMID: 27074859 DOI: 10.1049/iet-nbt.2015.0030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cell penetrating peptides (CPPs) were developed as vehicles for efficient delivery of various molecules. An ideal CPP-peptide should not display any toxicity against cancer cells as well as healthy cells and efficiently enter into the cell. Because of the cationic nature and the intrinsic vector capabilities, these peptides can cause cytotoxicity. One of the possible reasons for toxicity of CPPs is direct translocation and consequently, pore formation on the plasma membrane. In this study it was demonstrated that interaction of poly-glutamate with CPP considerably reduced their cytotoxicity in A549 cell. This strategy could be useful for efficient drug delivery mediated by CPP.
Collapse
Affiliation(s)
| | - Ali Shirani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Mohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Shahbazi Mojarrad
- Biotechnology Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Takechi-Haraya Y, Nadai R, Kimura H, Nishitsuji K, Uchimura K, Sakai-Kato K, Kawakami K, Shigenaga A, Kawakami T, Otaka A, Hojo H, Sakashita N, Saito H. Enthalpy-driven interactions with sulfated glycosaminoglycans promote cell membrane penetration of arginine peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:1339-49. [DOI: 10.1016/j.bbamem.2016.03.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/12/2016] [Accepted: 03/17/2016] [Indexed: 12/16/2022]
|
23
|
Grimaldi N, Andrade F, Segovia N, Ferrer-Tasies L, Sala S, Veciana J, Ventosa N. Lipid-based nanovesicles for nanomedicine. Chem Soc Rev 2016; 45:6520-6545. [DOI: 10.1039/c6cs00409a] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multifunctional lipid-based nanovesicles (L-NVs) prepared by molecular self-assembly of membrane components together with (bio)-active molecules, by means of compressed CO2-media or other non-conventional methods lead to highly homogeneous, tailor-made nanovesicles that are used for advanced nanomedicine. Confocal microscopy image of siRNA transfection using L-NVs, reprinted with permission from de Jonge,et al.,Gene Therapy, 2006,13, 400–411.
Collapse
Affiliation(s)
- N. Grimaldi
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Nanomol Technologies SA
| | - F. Andrade
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - N. Segovia
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - L. Ferrer-Tasies
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Nanomol Technologies SA
| | - S. Sala
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - J. Veciana
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| | - N. Ventosa
- Institut de Ciència de Materials de Barcelona (ICMAB-CSIC)
- Campus Universitari de Bellaterra
- Cerdanyola del Vallès
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
| |
Collapse
|
24
|
Abstract
In the era of biomedicines and engineered carrier systems, cell penetrating peptides (CPPs) have been established as a promising tool for therapeutic application. Likewise, other therapeutic peptides, successful in vivo application of CPPs will strongly depend on peptide stability, the bottleneck for this type of biodegradable molecules. In this review, the authors describe the current knowledge of the in vivo degradation for known CPPs and the different strategies available to provide a higher resistance to metabolic degradation while preserving cell penetration efficiency. Peptide stability can be improved by different means, either modifying the structure to make it unrecognizable to proteases, or preventing access of proteolytic enzymes by applying conformation restriction or shielding strategies.
Collapse
|
25
|
Moutal A, François-Moutal L, Brittain JM, Khanna M, Khanna R. Differential neuroprotective potential of CRMP2 peptide aptamers conjugated to cationic, hydrophobic, and amphipathic cell penetrating peptides. Front Cell Neurosci 2015; 8:471. [PMID: 25674050 PMCID: PMC4306314 DOI: 10.3389/fncel.2014.00471] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/30/2014] [Indexed: 01/26/2023] Open
Abstract
The microtubule-associated axonal specification collapsin response mediator protein 2 (CRMP2) is a novel target for neuroprotection. A CRMP2 peptide (TAT-CBD3) conjugated to the HIV transactivator of transcription (TAT) protein's cationic cell penetrating peptide (CPP) motif protected neurons in the face of toxic levels of Ca(2+) influx leaked in via N-methyl-D-aspartate receptor (NMDAR) hyperactivation. Here we tested whether replacing the hydrophilic TAT motif with alternative cationic (nona-arginine (R9)), hydrophobic (membrane transport sequence (MTS) of k-fibroblast growth factor) or amphipathic (model amphipathic peptide (MAP)) CPPs could be superior to the neuroprotection bestowed by TAT-CBD3. In giant plasma membrane vesicles (GPMVs) derived from cortical neurons, the peptides translocated across plasma membranes with similar efficiencies. Cortical neurons, acutely treated with peptides prior to a toxic glutamate challenge, demonstrated enhanced efflux of R9-CBD3 compared to others. R9-CBD3 inhibited N-methyl-D-aspartate (NMDA)-evoked Ca(2+) influx to a similar extent as TAT-CBD3 while MTS-CBD3 was ineffective which correlated with the ability of R9- and TAT-CBD3, but not MTS-CBD3, to block NMDAR interaction with CRMP2. Unrestricted Ca(2+) influx through NMDARs leading to delayed calcium dysregulation and neuronal cell death was blocked by all peptides but MAP-CBD3. When applied acutely for 10 min, R9-CBD3 was more effective than TAT-CBD3 at neuroprotection while MTS- and MAP-CBD3 were ineffective. In contrast, long-term (>24 h) treatment with MTS-CBD3 conferred neuroprotection where TAT-CBD3 failed. Neither peptide altered surface trafficking of NMDARs. Neuroprotection conferred by MTS-CBD3 peptide is likely due to its increased uptake coupled with decreased efflux when compared to TAT-CBD3. Overall, our results demonstrate that altering CPPs can bestow differential neuroprotective potential onto the CBD3 cargo.
Collapse
Affiliation(s)
- Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona Tucson, AZ, USA
| | | | - Joel M Brittain
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine Indianapolis, IN, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona Tucson, AZ, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona Tucson, AZ, USA ; Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona Tucson, AZ, USA
| |
Collapse
|
26
|
Gaspar V, de Melo-Diogo D, Costa E, Moreira A, Queiroz J, Pichon C, Correia I, Sousa F. Minicircle DNA vectors for gene therapy: advances and applications. Expert Opin Biol Ther 2014; 15:353-79. [PMID: 25539147 DOI: 10.1517/14712598.2015.996544] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Nucleic-acid-based biopharmaceuticals enclose a remarkable potential for treating debilitating or life-threatening diseases that currently remain incurable. This promising area of research envisages the creation of state-of-the-art DNA vaccines, pluripotent cells or gene-based therapies, which can be used to overcome current issues. To achieve this goal, DNA minicircles are emerging as ideal nonviral vectors due to their safety and persistent transgene expression in either quiescent or actively dividing cells. AREAS COVERED This review focuses on the characteristics of minicircle DNA (mcDNA) technology and the current advances in their production. The possible modifications to further improve minicircle efficacy are also emphasized and discussed in light of recent advances. As a final point, the main therapeutic applications of mcDNA are summarized, with a special focus on pluripotent stem cells production and cancer therapy. EXPERT OPINION Achieving in-target and persistent transgene expression is a challenging issue that is of critical importance for a successful therapeutic outcome. The use of miniaturized mcDNA cassettes with additional modifications that increase and prolong expression may contribute to an improved generation of biopharmaceuticals. The unique features of mcDNA render it an attractive alternative to overcome current technical issues and to bridge the significant gap that exists between basic research and clinical applications.
Collapse
Affiliation(s)
- Vítor Gaspar
- University of Beira Interior, CICS-UBI - Health Sciences Research Center , Av. Infante D. Henrique, 6200-506, Covilhã , Portugal +351 275 329 002, +351 275 329 055 ; +351 275 329 099 ; ;
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Tang J, Zhang L, Fu H, Kuang Q, Gao H, Zhang Z, He Q. A detachable coating of cholesterol-anchored PEG improves tumor targeting of cell-penetrating peptide-modified liposomes. Acta Pharm Sin B 2014; 4:67-73. [PMID: 26579366 PMCID: PMC4590295 DOI: 10.1016/j.apsb.2013.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/11/2013] [Accepted: 12/16/2013] [Indexed: 01/20/2023] Open
Abstract
Cell-penetrating peptides (CPPs) have been widely used to enhance the membrane translocation of various carriers for many years, but the non-specificity of CPPs seriously limits their utility in vivo. In this study, cholesterol-anchored, reduction-sensitive PEG (first synthesized by our laboratory) was applied to develop a co-modified liposome with improved tumor targeting. Following optimization of the formulation, the in vitro and in vivo properties of the co-modified liposome were evaluated. The co-modified liposome had a much lower cellular uptake and tumor spheroid uptake, but a much higher tumor accumulation compared to CPP-modified liposome, indicating the non-specific penetration of CPPs could be attenuated by the outer PEG coating. With the addition of exogenous reducing agent, both the in vitro and in vivo cellular uptake was markedly increased, demonstrating that the reduction-sensitive PEG coating achieved a controllable detachment from the surface of liposomes and did not affect the penetrating abilities of CPPs. The present results demonstrate that the combination of cholestervsitive PEG and CPPs is an ideal alternative for the application of CPP-modified carriers in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qin He
- Corresponding author at: West China School of Pharmacy, Sichuan University, No. 17 Block 3 Southern Renmin Road, Chengdu, Sichuan 610041, China. Tel./fax: +86 28 85502532.
| |
Collapse
|