1
|
Serebrennikova M, Grafskaia E, Maltsev D, Ivanova K, Bashkirov P, Kornilov F, Volynsky P, Efremov R, Bocharov E, Lazarev V. TriplEP-CPP: Algorithm for Predicting the Properties of Peptide Sequences. Int J Mol Sci 2024; 25:6869. [PMID: 38999985 PMCID: PMC11241344 DOI: 10.3390/ijms25136869] [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: 05/25/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Advancements in medicine and pharmacology have led to the development of systems that deliver biologically active molecules inside cells, increasing drug concentrations at target sites. This improves effectiveness and duration of action and reduces side effects on healthy tissues. Cell-penetrating peptides (CPPs) show promise in this area. While traditional medicinal chemistry methods have been used to develop CPPs, machine learning techniques can speed up and reduce costs in the search for new peptides. A predictive algorithm based on machine learning models was created to identify novel CPP sequences using molecular descriptors using a combination of algorithms like k-nearest neighbors, gradient boosting, and random forest. Some potential CPPs were found and tested for cytotoxicity and penetrating ability. A new low-toxicity CPP was discovered from the Rhopilema esculentum venom proteome through this study.
Collapse
Affiliation(s)
- Maria Serebrennikova
- Laboratory of Genetic Engineering, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (M.S.); (K.I.); (V.L.)
- Moscow Center for Advanced Studies 20, Kulakova Str., Moscow 123592, Russia; (P.B.); (F.K.); (R.E.); (E.B.)
| | - Ekaterina Grafskaia
- Laboratory of Genetic Engineering, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (M.S.); (K.I.); (V.L.)
| | - Dmitriy Maltsev
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia;
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Kseniya Ivanova
- Laboratory of Genetic Engineering, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (M.S.); (K.I.); (V.L.)
- Moscow Center for Advanced Studies 20, Kulakova Str., Moscow 123592, Russia; (P.B.); (F.K.); (R.E.); (E.B.)
- Research Institute for Systems Biology and Medicine, Moscow 117246, Russia
| | - Pavel Bashkirov
- Moscow Center for Advanced Studies 20, Kulakova Str., Moscow 123592, Russia; (P.B.); (F.K.); (R.E.); (E.B.)
- Research Institute for Systems Biology and Medicine, Moscow 117246, Russia
| | - Fedor Kornilov
- Moscow Center for Advanced Studies 20, Kulakova Str., Moscow 123592, Russia; (P.B.); (F.K.); (R.E.); (E.B.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
| | - Pavel Volynsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Roman Efremov
- Moscow Center for Advanced Studies 20, Kulakova Str., Moscow 123592, Russia; (P.B.); (F.K.); (R.E.); (E.B.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
| | - Eduard Bocharov
- Moscow Center for Advanced Studies 20, Kulakova Str., Moscow 123592, Russia; (P.B.); (F.K.); (R.E.); (E.B.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia;
| | - Vassili Lazarev
- Laboratory of Genetic Engineering, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow 119435, Russia; (M.S.); (K.I.); (V.L.)
- Moscow Center for Advanced Studies 20, Kulakova Str., Moscow 123592, Russia; (P.B.); (F.K.); (R.E.); (E.B.)
| |
Collapse
|
2
|
Zhai Y, Li S, Wang H, Shan Y. Revealing the dynamic mechanism of cell-penetrating peptides across cell membranes at the single-molecule level. J Mater Chem B 2024; 12:5589-5593. [PMID: 38741568 DOI: 10.1039/d4tb00522h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Cell-penetrating peptides (CPPs) have gained prominence in cellular drug delivery due to their extremely low toxicity and rapid cell internalization properties. Understanding the effect of CPPs' physicochemical properties on trans-membrane behavior will provide a better screening scheme for designing effective CPP-conjugated nano-drugs. Herein, the efficiency of the CPPs interacting with the cell membrane and the subsequent trans-membrane is revealed at the single-molecule level using single-molecule force spectroscopy (SMFS) and force tracing technique based on atomic force spectroscopy (AFM). The dynamic force spectroscopy (DFS) analysis indicates that cationic TAT48-60 and amphipathic MAP are more effective during the interaction with cell membrane due to the strong electrostatic interaction between CPPs and cell membrane. However, for the subsequent trans-membrane process, the hydrophobicity of Pep-7 plays a key role, showing a higher trans-membrane speed at the single-molecule level. Meanwhile, Pep-7 shows lower trans-membrane speed and probability on normal cells (Vero), which makes it more suitable as a peptide-based nano-drug to treat tumors avoiding harming normal cells. The dynamic parameters obtained in this study offer guidance for screening and modifying effective CPPs, targeting specific cell lines or tissues during the nano-drug design.
Collapse
Affiliation(s)
- Yuhang Zhai
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| | - Siying Li
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| | - Hui Wang
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| | - Yuping Shan
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| |
Collapse
|
3
|
Kotadiya DD, Patel P, Patel HD. Cell-Penetrating Peptides: A Powerful Tool for Targeted Drug Delivery. Curr Drug Deliv 2024; 21:368-388. [PMID: 37026498 DOI: 10.2174/1567201820666230407092924] [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: 09/27/2022] [Revised: 01/19/2023] [Accepted: 01/30/2023] [Indexed: 04/08/2023]
Abstract
The cellular membrane hinders the effective delivery of therapeutics to targeted sites. Cellpenetrating peptide (CPP) is one of the best options for rapidly internalizing across the cellular membrane. CPPs have recently attracted lots of attention because of their excellent transduction efficiency and low cytotoxicity. The CPP-cargo complex is an effective and efficient method of delivering several chemotherapeutic agents used to treat various diseases. Additionally, CPP has become another strategy to overcome some of the current therapeutic agents' limitations. However, no CPP complex is approved by the US FDA because of its limitations and issues. In this review, we mainly discuss the cellpenetrating peptide as the delivery vehicle, the cellular uptake mechanism of CPPs, their design, and some strategies to synthesize the CPP complex via some linkers such as disulfide bond, oxime, etc. Here, we also discuss the recent status of CPPs in the market.
Collapse
Affiliation(s)
- Dushyant D Kotadiya
- Department of Chemistry, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Piyushkumar Patel
- Department of Chemistry, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| | - Hitesh D Patel
- Department of Chemistry, School of Sciences, Gujarat University, Ahmedabad, 380009, Gujarat, India
| |
Collapse
|
4
|
Improved prediction and characterization of blood-brain barrier penetrating peptides using estimated propensity scores of dipeptides. J Comput Aided Mol Des 2022; 36:781-796. [DOI: 10.1007/s10822-022-00476-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/15/2022] [Indexed: 11/27/2022]
|
5
|
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
|
6
|
Rusiecka I, Gągało I, Kocić I. Cell-penetrating peptides improve pharmacokinetics and pharmacodynamics of anticancer drugs. Tissue Barriers 2022; 10:1965418. [PMID: 34402743 PMCID: PMC8794253 DOI: 10.1080/21688370.2021.1965418] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022] Open
Abstract
This review concentrates on the research concerning conjugates of anticancer drugs with versatile cell-penetrating peptides (CPPs). For a better insight into the relationship between the components of the constructs, it starts with the characteristic of the peptides and considers its following aspects: mechanisms of cellular internalization, interaction with cancer-modified membranes, selectivity against tumor tissue. Also, CPPs with anticancer activity have been distinguished and summarized with their mechanisms of action. With respect to the conjugates, the preclinical studies (in vitro, in vivo) indicated that they possess several merits in comparison to the parent drugs. They concerned not only better cellular internalization but also other improvements in pharmacokinetics (e.g. access to the brain tissue) and pharmacodynamics (e.g. overcoming drug resistance). The anticancer activity of the conjugates was usually superior to that of the unconjugated drug. Certain anticancer CPPs and conjugates entered clinical trials.
Collapse
Affiliation(s)
- Izabela Rusiecka
- Department of Pharmacology, Medical University of Gdansk, Gdansk, Poland
| | - Iwona Gągało
- Department of Pharmacology, Medical University of Gdansk, Gdansk, Poland
| | - Ivan Kocić
- Department of Pharmacology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
7
|
In Vitro and In Vivo Studies on the Antibacterial Activity and Safety of a New Antimicrobial Peptide Dermaseptin-AC. Microbiol Spectr 2021; 9:e0131821. [PMID: 34908502 PMCID: PMC8672897 DOI: 10.1128/spectrum.01318-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Antimicrobial resistance has been an increasing public health threat in recent years. Antimicrobial peptides are considered as potential drugs against drug-resistant bacteria because they are mainly broad-spectrum and are unlikely to cause resistance. In this study, a novel peptide was obtained from the skin secretion of Agalychnis callidryas using the “shotgun” cloning method. The amino acid sequence, molecular weight, and secondary structure of Dermaseptin-AC were determined. The in vitro antimicrobial activity, hemolysis, and cytotoxicity of Dermaseptin-AC were evaluated. MICs and minimum bactericidal concentrations (MBCs) of Dermaseptin-AC against seven different bacterial strains ranged between 2 ∼ 4 μM and 2 ∼ 8 μM. The HC50 (50% maximum hemolysis concentration) of Dermaseptin-AC against horse erythrocytes was 76.55 μM. The in vivo anti-MRSA effect was tested on immune-suppressed MRSA pneumonia in mice. Dermaseptin-AC showed anti-MRSA effects similar to the same dose of vancomycin (10 mg/kg body weight). Short-term (7 days of intraperitoneal injection, 10 mg/kg body weight) in vivo safety evaluation of Dermaseptin-AC was tested on mice. The survival rate during the 7-day injection was 80%. Dermaseptin-AC showed no obvious effect on the liver, heart, spleen, kidney, and blood, but did induce slight pulmonary congestion. The skin safety of Dermaseptin-AC was evaluated on wounds on the back skin of a rat, and no irritation was observed. IMPORTANCE In this study, we discovered a new antimicrobial peptide, Dermaseptin-AC, and studied its in vitro and in vivo antimicrobial activity. These studies provide some data for finding new antimicrobial peptides for overcoming antimicrobial resistance. Dermaseptin-AC showed strong broad-spectrum antibacterial activity and relatively low hemolysis, and was more cytotoxic to cancer cells than to normal cells. Dermaseptin-AC was active in vivo, and its anti-MRSA effect was similar to that of vancomycin when administered by intraperitoneal injection. Safety studies found that continuous injection of Dermaseptin-AC may cause mild pulmonary congestion, while there was no obvious irritation when it was applied to skin wounds. Chronic wounds are often accompanied by high bacterial burdens and, at the same time, antimicrobial resistance is more likely to occur during repeated infections and treatments. Therefore, developing Dermaseptin-AC to treat chronic wound infection may be an attractive choice.
Collapse
|
8
|
Abstract
Cell penetrating peptides (CPPs) are short peptides that are able to translocate themselves and their cargo into cells. The progressive and continuous application of CPPs in various fields of basic and applied research shows that they are efficient delivery vectors for an assortment of biomolecules, including nucleic acids and proteins. This feature makes CPPs an excellent tool for modification of plant genomes through transgenesis and genome editing. In this review, we present the progress during the last three decades in application of CPPs for delivery of DNA, RNA, and proteins into plant cells and tissues. Moreover, we highlight the exploiting of CPPs as advantageous and beneficial tool for plant genome editing via delivery of nuclease proteins, and provide a practical example of genome alternation through CPP-delivered nucleases. Finally, the current exploitation of peptides in organelle-specific DNA delivery and modification of organellar genomes is discussed.
Collapse
|
9
|
Tarvirdipour S, Skowicki M, Schoenenberger CA, Palivan CG. Peptide-Assisted Nucleic Acid Delivery Systems on the Rise. Int J Mol Sci 2021; 22:9092. [PMID: 34445799 PMCID: PMC8396486 DOI: 10.3390/ijms22169092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Concerns associated with nanocarriers' therapeutic efficacy and side effects have led to the development of strategies to advance them into targeted and responsive delivery systems. Owing to their bioactivity and biocompatibility, peptides play a key role in these strategies and, thus, have been extensively studied in nanomedicine. Peptide-based nanocarriers, in particular, have burgeoned with advances in purely peptidic structures and in combinations of peptides, both native and modified, with polymers, lipids, and inorganic nanoparticles. In this review, we summarize advances on peptides promoting gene delivery systems. The efficacy of nucleic acid therapies largely depends on cell internalization and the delivery to subcellular organelles. Hence, the review focuses on nanocarriers where peptides are pivotal in ferrying nucleic acids to their site of action, with a special emphasis on peptides that assist anionic, water-soluble nucleic acids in crossing the membrane barriers they encounter on their way to efficient function. In a second part, we address how peptides advance nanoassembly delivery tools, such that they navigate delivery barriers and release their nucleic acid cargo at specific sites in a controlled fashion.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- Department of Biosystem Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Michal Skowicki
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| |
Collapse
|
10
|
Abstract
Cell-Penetrating Peptides (CPP) are valuable tools capable of crossing the plasma membrane to deliver therapeutic cargo inside cells. Small interfering RNAs (siRNA) are double-stranded RNA molecules capable of silencing the expression of a specific protein triggering the RNA interference (RNAi) pathway, but they are unable to cross the plasma membrane and have a short half-life in the bloodstream. In this overview, we assessed the many different approaches used and developed in the last two decades to deliver siRNA through the plasma membrane through different CPPs sorted according to three different loading strategies: covalent conjugation, complex formation, and CPP-decorated (functionalized) nanocomplexes. Each of these strategies has pros and cons, but it appears the latter two are the most commonly reported and emerging as the most promising strategies due to their simplicity of synthesis, use, and versatility. Recent progress with siRNA delivered by CPPs seems to focus on targeted delivery to reduce side effects and amount of drugs used, and it appears to be among the most promising use for CPPs in future clinical applications.
Collapse
|
11
|
Asfour MH. Advanced trends in protein and peptide drug delivery: a special emphasis on aquasomes and microneedles techniques. Drug Deliv Transl Res 2020; 11:1-23. [PMID: 32337668 DOI: 10.1007/s13346-020-00746-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proteins and peptides have a great potential as therapeutic agents; they have higher efficiency and lower toxicity, compared to chemical drugs. However, their oral bioavailability is very low; also, the transdermal peptide delivery faces absorption limitations. Accordingly, most of proteins and peptides are administered by parenteral route, but there are many problems associated with this route such as patient discomfort, especially for pediatric use. Thus, it is a great challenge to develop drug delivery systems for administration of proteins and peptides by routes other than parenteral one. This review provides an overview on recent advances adopted for protein and peptide drug delivery, focusing on oral and transdermal routes. This is followed by an emphasis on two recent approaches adopted as delivery systems for protein and peptide drugs, namely aquasomes and microneedles. Aquasomes are nanoparticles fabricated from ceramics developed to enhance proteins and peptides stability, providing an adequate residence time in circulation. It consists of ceramic core coated with poly hydroxyl oligomer, on which protein and peptide drug can be adsorbed. Aquasomes preparation, characterization, and application in protein and peptide drug delivery are discussed. Microneedles are promising transdermal approach; it involves creation of micron-sized pores in the skin for enhancing the drug delivery across the skin, as their length ranged between 150 and 1500 μm. Types of microneedles with different drug delivery mechanisms, characterization, and application in protein and peptide drug delivery are discussed. Graphical abstract.
Collapse
Affiliation(s)
- Marwa Hasanein Asfour
- Pharmaceutical Technology Department, National Research Centre, El-Buhouth Street, Dokki, Cairo, 12622, Egypt.
| |
Collapse
|
12
|
Zarei M, Rahbar MR, Negahdaripour M, Morowvat MH, Nezafat N, Ghasemi Y. Cell Penetrating Peptide: Sequence-Based Computational Prediction for Intercellular Delivery of Arginine Deiminase. CURR PROTEOMICS 2020. [DOI: 10.2174/1570164616666190701120351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:Cell-Penetrating Peptides (CPPs), a family of short peptides, are broadly used as the carrier in the delivery of drugs and different therapeutic agents. Thanks to the existence of valuable databases, computational screening of the experimentally validated CPPs can help the researchers to select more effective CPPs for the intercellular delivery of therapeutic proteins. Arginine deiminase of Mycoplasma hominis, an arginine-degrading enzyme, is currently in the clinical trial for treating several arginine auxotrophic cancers. However, some tumor cells have developed resistance to ADI treatment. The ADI resistance arises from the over-expression of argininosuccinate synthetase 1 enzyme, which is involved in arginine synthesis. Intracellular delivery of ADI into tumor cells is suggested as an efficient approach to overcome the aforesaid drawback.Objective:In this study, in-silico tools were used for evaluating the experimentally validated CPPs to select the best CPP candidates for the intracellular delivery of ADI.Results:In this regard, 150 CPPs of protein cargo available at CPPsite were retrieved and evaluated by the CellPPD server. The best CPP candidates for the intracellular delivery of ADI were selected based on stability and antigenicity of the ADI-CPP fusion form. The conjugated forms of ADI with each of the three CPPs including EGFP-hcT (9-32), EGFP-ppTG20, and F(SG)4TP10 were stable and nonantigenic; thus, these sequences were introduced as the best CPP candidates for the intracellular delivery of ADI. In addition, the proposed CPPs had appropriate positive charge and lengths for an efficient cellular uptake.Conclusion:These three introduced CPPs not only are appropriate for the intracellular delivery of ADI, but also can overcome the limitation of its therapeutic application, including short half-life and antigenicity.
Collapse
Affiliation(s)
- Mahboubeh Zarei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Rahbar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Thotakura N, Kaushik L, Kumar V, Preet S, Babu PV. Advanced Approaches of Bioactive Peptide Molecules and Protein Drug Delivery Systems. Curr Pharm Des 2019; 24:5147-5163. [PMID: 30727874 DOI: 10.2174/1381612825666190206211458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/01/2019] [Indexed: 11/22/2022]
Abstract
Despite the fact that protein and peptide therapeutics are widely employed in the treatment of various diseases, their delivery is posing an unembellished challenge to the scientists. It was discovered that delivery of these therapeutic systems through oral route is easy with high patient compliance. However, proteolytic degradation and absorption through the mucosal epithelium are the barriers in this route. These issues can be minimized by the use of enzyme inhibitors, absorption enhancers, different carrier systems or either by direct modification. In the process of investigation, it was found that transdermal route is not posing any challenges of enzymatic degradation, but, still absorption is the limitation as the outer layer of skin acts as a barrier. To suppress the effect of the barrier and increase the rate of the absorption, various advanced technologies were developed, namely, microneedle technology, iontophoresis, electroporation, sonophoresis and biochemical enhancement. Indeed, even these molecules are targeted to the cells with the use of cell-penetrating peptides. In this review, delivery of the peptide and protein therapeutics using oral, transdermal and other routes is discussed in detail.
Collapse
Affiliation(s)
- Nagarani Thotakura
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan, India
| | - Lokesh Kaushik
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan, India
| | - Vipin Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Ajmer, Rajasthan, India
| | - Simran Preet
- Department of Biophysics, Basic Medical Sciences Block-2, Panjab University, Sector-25, Chandigarh, India
| | - Penke Vijaya Babu
- Department of chemistry, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
14
|
Toyohara D, Yokoi Y, Inoue G, Muraoka T, Mori T. Abiotic Factors Promote Cell Penetrating Peptide Permeability in Enterobacteriaceae Models. Front Microbiol 2019; 10:2534. [PMID: 31849846 PMCID: PMC6902036 DOI: 10.3389/fmicb.2019.02534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/21/2019] [Indexed: 11/13/2022] Open
Abstract
Conventionally, the delivery of biomolecules into bacteria for the generation of characterized or functional mutants has relied greatly on horizontal gene transfer techniques. However, the low compatibility of these techniques with novel or hard-to-transform bacteria currently serves as a challenge to the bioengineering field. Here, we explored the use of cell penetrating peptides (CPPs) as an alternative biomolecule delivery approach by investigating the effects of the abiotic factors during CPP permeation. Using the (KFF)3K-FAM conjugate and Escherichia coli as models, we evaluated four abiotic factors where two of these factors, temperature and solution tonicity, promoted (KFF)3K-FAM permeation efficiency. Our data show that optimal (KFF)3K-FAM permeation efficiency was achieved for E. coli at approximately 98.1% under conditions of 37°C (growth optimal temperature) and 50% PBS concentration. Based on these conditions, we subsequently tested the applicability of CPP permeation in various bacterial strains by treating 10 bacterial strains from the Enterobacteriaceae family among which seven strains have no CPP permeation records with (KFF)3K-FAM. Interestingly, when compared with non-optimized conditions, all 10 strains showed a marked increase in CPP permeation ranging between 20 and 90% efficiency. Although using strains within Enterobacteriaceae that are phylogenetically close, our results hinted on the possibility that with proper optimization of the abiotic factors, CPPs could be compatible with a broad range of bacterial strains. Our efforts suggest that CPP could serve as an effective alternative approach for mutant generation and for biomolecule delivery into novel or hard-to-transform bacteria.
Collapse
Affiliation(s)
- Daichi Toyohara
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Japan
| | - Yasuhito Yokoi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Japan
| | - Go Inoue
- Department of Organic and Polymer Materials Chemistry, Tokyo University of Agriculture and Technology, Koganei, Japan
| | - Takahiro Muraoka
- Department of Organic and Polymer Materials Chemistry, Tokyo University of Agriculture and Technology, Koganei, Japan
| | - Tetsushi Mori
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Koganei, Japan
| |
Collapse
|
15
|
Schiroli D, Gómara MJ, Maurizi E, Atkinson SD, Mairs L, Christie KA, Cobice DF, McCrudden CM, Nesbit MA, Haro I, Moore T. Effective In Vivo Topical Delivery of siRNA and Gene Silencing in Intact Corneal Epithelium Using a Modified Cell-Penetrating Peptide. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:891-906. [PMID: 31476668 PMCID: PMC6723413 DOI: 10.1016/j.omtn.2019.07.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 01/03/2023]
Abstract
Autosomal dominantly inherited genetic disorders such as corneal dystrophies are amenable to allele-specific gene silencing with small interfering RNA (siRNA). siRNA delivered to the cornea by injection, although effective, is not suitable for a frequent long-term treatment regimen, whereas topical delivery of siRNA to the cornea is hampered by the eye surface's protective mechanisms. Herein we describe an attractive and innovative alternative for topical application using cell-penetrating peptide derivatives capable of complexing siRNA non-covalently and delivering them into the cornea. Through a rational design approach, we modified derivatives of a cell-penetrating peptide, peptide for ocular delivery (POD), already proved to diffuse into the corneal layers. These POD derivatives were able to form siRNA-peptide complexes (polyplexes) of size and ζ-potential similar to those reported able to undergo cellular internalization. Successful cytoplasmic release and gene silencing in vitro was obtained when an endosomal disruptor, chloroquine, was added. A palmitoylated-POD, displaying the best delivery properties, was covalently functionalized with trifluoromethylquinoline, an analog of chloroquine. This modified POD, named trifluoromethylquinoline-palmitoyl-POD (QN-Palm-POD), when complexed with siRNA and topically applied to the eye in vivo, resulted in up to 30% knockdown of luciferase reporter gene expression in the corneal epithelium. The methods developed within represent a valid standardized approach that is ideal for screening of a range of delivery formulations.
Collapse
Affiliation(s)
- Davide Schiroli
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland
| | - María J Gómara
- Unit of Synthesis and Biomedical Applications of Peptides, Department of Biomedical Chemistry, Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - Eleonora Maurizi
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland
| | - Sarah D Atkinson
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland; Northern Ireland Centre for Stratified Medicine, University of Ulster, Londonderry BT47 6SB, UK
| | - Laura Mairs
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland
| | - Kathleen A Christie
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland
| | - Diego F Cobice
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland
| | - Cian M McCrudden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland
| | - M Andrew Nesbit
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland
| | - Isabel Haro
- Unit of Synthesis and Biomedical Applications of Peptides, Department of Biomedical Chemistry, Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - Tara Moore
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, Northern Ireland.
| |
Collapse
|
16
|
Deprey K, Becker L, Kritzer J, Plückthun A. Trapped! A Critical Evaluation of Methods for Measuring Total Cellular Uptake versus Cytosolic Localization. Bioconjug Chem 2019; 30:1006-1027. [PMID: 30882208 PMCID: PMC6527423 DOI: 10.1021/acs.bioconjchem.9b00112] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biomolecules have many properties that make them promising for intracellular therapeutic applications, but delivery remains a key challenge because large biomolecules cannot easily enter the cytosol. Furthermore, quantification of total intracellular versus cytosolic concentrations remains demanding, and the determination of delivery efficiency is thus not straightforward. In this review, we discuss strategies for delivering biomolecules into the cytosol and briefly summarize the mechanisms of uptake for these systems. We then describe commonly used methods to measure total cellular uptake and, more selectively, cytosolic localization, and discuss the major advantages and drawbacks of each method. We critically evaluate methods of measuring "cell penetration" that do not adequately distinguish total cellular uptake and cytosolic localization, which often lead to inaccurate interpretations of a molecule's cytosolic localization. Finally, we summarize the properties and components of each method, including the main caveats of each, to allow for informed decisions about method selection for specific applications. When applied correctly and interpreted carefully, methods for quantifying cytosolic localization offer valuable insight into the bioactivity of biomolecules and potentially the prospects for their eventual development into therapeutics.
Collapse
Affiliation(s)
- Kirsten Deprey
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford, Massachusetts 02155, United States
| | - Lukas Becker
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Joshua Kritzer
- Department of Chemistry, Tufts University, 62 Talbot Avenue, Medford, Massachusetts 02155, United States
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
17
|
Lin CC, Bachmann M, Bachler S, Venkatesan K, Dittrich PS. Tunable Membrane Potential Reconstituted in Giant Vesicles Promotes Permeation of Cationic Peptides at Nanomolar Concentrations. ACS APPLIED MATERIALS & INTERFACES 2018; 10:41909-41916. [PMID: 30450894 PMCID: PMC6420060 DOI: 10.1021/acsami.8b12217] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
We investigate the influence of membrane potential on the permeation of cationic peptides. Therefore, we employ a microfluidic chip capable of capturing giant unilamellar vesicles (GUVs) in physical traps and fast exchange of chemical compounds. Control experiments with calcein proved that the vesicle membranes' integrity is not affected by the physical traps and applied shear forces. Combined with fluorescence correlation spectroscopy, permeation of fluorescently labeled peptides across vesicle membranes can be measured down to the nanomolar level. With the addition of a lipophilic ruthenium(II) complex Ru(C17)22+, GUVs consisting of mixed acyl phospholipids are prepared with a negative membrane potential, resembling the membrane asymmetry in cells. The membrane potential serves as a driving force for the permeation of cationic cell-penetrating peptides (CPPs) nonaarginine (Arg9) and the human immunodeficiency virus trans-activator of transcription (TAT) peptide already at nanomolar doses. Hyperpolarization of the membrane by photo-oxidation of Ru(C17)22+ enhances permeation significantly from 55 to 78% for Arg9. This specific enhancement for Arg9 (cf. TAT) is ascribed to the higher affinity of the arginines to the phosphoserine head groups. On the other hand, permeation is decreased by introducing an additional negative charge in close proximity to the N-terminal arginine residue when changing the fluorophore. In short, with the capability to reconstitute membrane potential as well as shear stress, our system is a suitable platform for modeling the membrane permeability of pharmaceutics candidates. The results also highlight the membrane potential as a major cause of discrepancies between vesicular and cellular studies on CPP permeation.
Collapse
Affiliation(s)
- Chao-Chen Lin
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Michael Bachmann
- Department of Chemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Simon Bachler
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Koushik Venkatesan
- Department of Chemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
- Department of Molecular Sciences, Macquarie University, North Ryde, NSW 2109, Australia
| | - Petra S. Dittrich
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| |
Collapse
|
18
|
A platform for discovery of functional cell-penetrating peptides for efficient multi-cargo intracellular delivery. Sci Rep 2018; 8:12538. [PMID: 30135446 PMCID: PMC6105642 DOI: 10.1038/s41598-018-30790-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022] Open
Abstract
Cell penetrating peptides (CPPs) offer great potential to deliver therapeutic molecules to previously inaccessible intracellular targets. However, many CPPs are inefficient and often leave their attached cargo stranded in the cell’s endosome. We report a versatile platform for the isolation of peptides delivering a wide range of cargos into the cytoplasm of cells. We used this screening platform to identify multiple “Phylomer” CPPs, derived from bacterial and viral genomes. These peptides are amenable to conventional sequence optimization and engineering approaches for cell targeting and half-life extension. We demonstrate potent, functional delivery of protein, peptide, and nucleic acid analog cargos into cells using Phylomer CPPs. We validate in vivo activity in the cytoplasm, through successful transport of an oligonucleotide therapeutic fused to a Phylomer CPP in a disease model for Duchenne’s muscular dystrophy. This report thus establishes a discovery platform for identifying novel, functional CPPs to expand the delivery landscape of druggable intracellular targets for biological therapeutics.
Collapse
|
19
|
Van der Poorten O, Legrand B, Vezenkov LL, García-Pindado J, Bettache N, Knuhtsen A, Pedersen DS, Sánchez-Navarro M, Martinez J, Teixidó M, Garcia M, Tourwé D, Amblard M, Ballet S. Indoloazepinone-Constrained Oligomers as Cell-Penetrating and Blood-Brain-Barrier-Permeating Compounds. Chembiochem 2018; 19:696-705. [PMID: 29377388 DOI: 10.1002/cbic.201700678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Indexed: 12/29/2022]
Abstract
Non-cationic and amphipathic indoloazepinone-constrained (Aia) oligomers have been synthesized as new vectors for intracellular delivery. The conformational preferences of the [l-Aia-Xxx]n oligomers were investigated by circular dichroism (CD) and NMR spectroscopy. Whereas Boc-[l-Aia-Gly]2,4 -OBn oligomers 12 and 13 and Boc-[l-Aia-β3 -h-l-Ala]2,4 -OBn oligomers 16 and 17 were totally or partially disordered, Boc-[l-Aia-l-Ala]2 -OBn (14) induced a typical turn stabilized by C5 - and C7 -membered H-bond pseudo-cycles and aromatic interactions. Boc-[l-Aia-l-Ala]4 -OBn (15) exhibited a unique structure with remarkable T-shaped π-stacking interactions involving the indole rings of the four l-Aia residues forming a dense hydrophobic cluster. All of the proposed FITC-6-Ahx-[l-Aia-Xxx]4 -NH2 oligomers 19-23, with the exception of FITC-6-Ahx-[l-Aia-Gly]4 -NH2 (18), were internalized by MDA-MB-231 cells with higher efficiency than the positive references penetratin and Arg8 . In parallel, the compounds of this series were successfully explored in an in vitro blood-brain barrier (BBB) permeation assay. Although no passive diffusion permeability was observed for any of the tested Ac-[l-Aia-Xxx]4 -NH2 oligomers in the PAMPA model, Ac-[l-Aia-l-Arg]4 -NH2 (26) showed significant permeation in the in vitro cell-based human model of the BBB, suggesting an active mechanism of cell penetration.
Collapse
Affiliation(s)
- Olivier Van der Poorten
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Baptiste Legrand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Lubomir L Vezenkov
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Júlia García-Pindado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Nadir Bettache
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Astrid Knuhtsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100, Copenhagen, Denmark
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100, Copenhagen, Denmark
| | - Macarena Sánchez-Navarro
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Marcel Garcia
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Muriel Amblard
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 5, France
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| |
Collapse
|
20
|
Wang H, Ma JL, Yang YG, Song Y, Wu J, Qin YY, Zhao XL, Wang J, Zou LL, Wu JF, Li JM, Liu CB. Efficient therapeutic delivery by a novel cell-permeant peptide derived from KDM4A protein for antitumor and antifibrosis. Oncotarget 2018; 7:49075-49090. [PMID: 27081693 PMCID: PMC5226491 DOI: 10.18632/oncotarget.8682] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/28/2016] [Indexed: 01/23/2023] Open
Abstract
Cell-penetrating peptide (CPP) based delivery have provided immense potential for the therapeutic applications, however, most of nonhuman originated CPPs carry the risk of possible cytotoxicity and immunogenicity, thus may restricting to be used. Here, we describe a novel human-derived CPP, denoted hPP10, and hPP10 has cell-penetrating properties evaluated by CellPPD web server, as well as In-Vitro and In-Vivo analysis. In vitro studies showed that hPP10-FITC was able to penetrate into various cells including primary cultured cells, likely through an endocytosis pathway. And functionalized macromolecules, such as green fluorescent protein (GFP), tumor-specific apoptosis inducer Apoptin as well as biological active enzyme GCLC (Glutamate-cysteine ligase, catalytic subunit) can be delivered by hPP10 in vitro and in vivo. Collectively, our results suggest that hPP10 provide a novel and versatile tool to deliver exogenous proteins or drugs for clinical applications as well as reprogrammed cell-based therapy.
Collapse
Affiliation(s)
- Hu Wang
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,Medical School, China Three Gorges University, Yichang 443002, China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Jie-Lan Ma
- Medical School, China Three Gorges University, Yichang 443002, China
| | - Ying-Gui Yang
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,Medical School, China Three Gorges University, Yichang 443002, China
| | - Yang Song
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,Medical School, China Three Gorges University, Yichang 443002, China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Jiao Wu
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,Medical School, China Three Gorges University, Yichang 443002, China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Yan-Yan Qin
- Medical School, China Three Gorges University, Yichang 443002, China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Xue-Li Zhao
- Medical School, China Three Gorges University, Yichang 443002, China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Jun Wang
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,The 1st People's Hospital of Yichang, Yichang 443000, China
| | - Li-Li Zou
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,Medical School, China Three Gorges University, Yichang 443002, China
| | - Jiang-Feng Wu
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,Medical School, China Three Gorges University, Yichang 443002, China
| | - Jun-Ming Li
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,The 1st People's Hospital of Yichang, Yichang 443000, China
| | - Chang-Bai Liu
- The Institute of Cell Therapy, China Three Gorges University, Yichang 443002, China.,Medical School, China Three Gorges University, Yichang 443002, China.,Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
21
|
Chaulagain B, Jain A, Tiwari A, Verma A, Jain SK. Passive delivery of protein drugs through transdermal route. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:472-487. [PMID: 29378433 DOI: 10.1080/21691401.2018.1430695] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Skin is the largest external organ in the human body but its use for therapeutic purposes has been minimal. Stratum corneum residing on the uppermost layer of the skin provides a tough barrier to transport the drugs across the skin. Very small group of drugs sharing Lipinski properties, i.e. drugs having molecular weight not larger than 500 Da, having high lipophilicity and optimum polarity are fortunate enough to be used on skin therapeutics. But, at a time where modern therapeutics is slowly shifting from use of small molecular drugs towards the use of macromolecular therapeutic agents such as peptides, proteins and nucleotides in origin, skin therapeutics need to be evolved accordingly to cater the delivery of these agents. Physical technologies like iontophoresis, laser ablation, micro-needles and ultrasound, etc. have been introduced to enhance skin permeability. But their success is limited due to their complex working mechanisms and involvement of certain irreversible skin damage in some or other way. This review therefore explores the delivery strategies for transport of mainly peptide and protein drugs that do not involve any injuries (non-invasive) to the skin termed as passive delivery techniques. Chemical enhancers, nanocarriers, certain biological peptides and miscellaneous approaches like prodrugs are also thoroughly reviewed for their applications in protein delivery.
Collapse
Affiliation(s)
- Bivek Chaulagain
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| | - Ankit Jain
- b Institute of Pharmaceutical Research, GLA University , Mathura , India
| | - Ankita Tiwari
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| | - Amit Verma
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| | - Sanjay K Jain
- a Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory , Dr. Hari Singh Gour Central University , Sagar , India
| |
Collapse
|
22
|
Park CK, Kim YH, Hwangbo S, Cho H. Photodynamic therapy by conjugation of cell-penetrating peptide with fluorochrome. Int J Nanomedicine 2017; 12:8185-8196. [PMID: 29184407 PMCID: PMC5689026 DOI: 10.2147/ijn.s148332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising alternative therapy that could be used as an adjunct to chemotherapy and surgery for cancer, and works by destroying tissue with visible light in the presence of a photosensitizer (PS) and oxygen. The PS should restrict tissue destruction only to the tumor and be activated by light of a specific wavelength; both of these properties are required. Arginine-rich peptides, such as cell-penetrating peptides, have membrane-translocating and nuclear-localizing activities, which have led to their application in various drug delivery modalities. Protamine (Pro) is an arginine-rich peptide with membrane-translocating and nuclear-localizing properties. The reaction of an N-hydroxysuccinimide (NHS) ester of rhodamine (Rho) and clinical Pro was carried out in this study to yield RhoPro, and a demonstration of its phototoxicity, wherein clinical Pro improved the effect of PDT, was performed. The reaction between Pro and the NHS ester of Rho is a solution-phase reaction that results in the complete modification of the Pro peptides, which feature a single reactive amine at the N-terminal proline and a single carboxyl group at the C-terminal arginine. This study aimed to identify a new type of PS for PDT by in vitro and in vivo experiments and to assess the antitumor effects of PDT, using the Pro-conjugated PS, on a cancer cell line. Photodynamic cell death studies showed that the RhoPro produced has more efficient photodynamic activities than Rho alone, causing rapid light-induced cell death. The attachment of clinical Pro to Rho, yielding RhoPro, confers the membrane-internalizing activity of its arginine-rich content on the fluorochrome Rho and can induce rapid photodynamic cell death, presumably owing to light-induced cell membrane rupture. PDT using RhoPro for HT-29 cells was very effective and these findings suggest that RhoPro is a suitable candidate as a PS for solid tumors.
Collapse
Affiliation(s)
- Chul-Kyu Park
- Department of Physiology, College of Medicine, Gachon University, Incheon
| | - Yong Ho Kim
- Department of Physiology, College of Medicine, Gachon University, Incheon
| | - Suhyun Hwangbo
- School of Materials Science & Engineering, Chonnam National University, Gwangju, South Korea
| | - Hoonsung Cho
- School of Materials Science & Engineering, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
23
|
Cell-Penetrating Peptides: Design Strategies beyond Primary Structure and Amphipathicity. Molecules 2017; 22:molecules22111929. [PMID: 29117144 PMCID: PMC6150340 DOI: 10.3390/molecules22111929] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/31/2017] [Accepted: 11/04/2017] [Indexed: 12/21/2022] Open
Abstract
Efficient intracellular drug delivery and target specificity are often hampered by the presence of biological barriers. Thus, compounds that efficiently cross cell membranes are the key to improving the therapeutic value and on-target specificity of non-permeable drugs. The discovery of cell-penetrating peptides (CPPs) and the early design approaches through mimicking the natural penetration domains used by viruses have led to greater efficiency of intracellular delivery. Following these nature-inspired examples, a number of rationally designed CPPs has been developed. In this review, a variety of CPP designs will be described, including linear and flexible, positively charged and often amphipathic CPPs, and more rigid versions comprising cyclic, stapled, or dimeric and/or multivalent, self-assembled peptides or peptido-mimetics. The application of distinct design strategies to known physico-chemical properties of CPPs offers the opportunity to improve their penetration efficiency and/or internalization kinetics. This led to increased design complexity of new CPPs that does not always result in greater CPP activity. Therefore, the transition of CPPs to a clinical setting remains a challenge also due to the concomitant involvement of various internalization routes and heterogeneity of cells used in the in vitro studies.
Collapse
|
24
|
A New Noncanonical Anionic Peptide That Translocates a Cellular Blood-Brain Barrier Model. Molecules 2017; 22:molecules22101753. [PMID: 29057814 PMCID: PMC6151732 DOI: 10.3390/molecules22101753] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/14/2017] [Indexed: 12/31/2022] Open
Abstract
The capacity to transport therapeutic molecules across the blood–brain barrier (BBB) represents a breakthrough in the development of tools for the treatment of many central nervous system (CNS)-associated diseases. The BBB, while being protective against infectious agents, hinders the brain uptake of many drugs. Hence, finding safe shuttles able to overcome the BBB is of utmost importance. Herein, we identify a new BBB-translocating peptide with unique properties. For years it was thought that cationic sequences were mandatory for a cell-penetrating peptide (CPP) to achieve cellular internalization. Despite being anionic at physiological pH, PepNeg (sequence (SGTQEEY) is an efficient BBB translocator that is able to carry a large cargo (27 kDa), while maintaining BBB integrity. In addition, PepNeg is able to use two distinct methods of translocation, energy-dependent and -independent, suggesting that direct penetration might occur when low concentrations of peptide are presented to cells. The discovery of this new anionic trans-BBB peptide allows the development of new delivery systems to the CNS and contributes to the need to rethink the role of electrostatic attraction in BBB-translocation.
Collapse
|
25
|
Mohammadi S, Zakeri-Milani P, Golkar N, Farkhani SM, Shirani A, Shahbazi Mojarrad J, Nokhodchi A, Valizadeh H. Synthesis and cellular characterization of various nano-assemblies of cell penetrating peptide-epirubicin-polyglutamate conjugates for the enhancement of antitumor activity. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1572-1585. [PMID: 28933182 DOI: 10.1080/21691401.2017.1379016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A new class of cell penetrating peptides (CPPs) named peptide amphiphile was designed to improve the intracellular uptake and the antitumor activity of epirubicin (EPR). Various amphiphilic CPPs were synthesized by solid phase peptide synthesis method and were chemically conjugated to EPR. Their corresponding nanoparticles (CPPs-E4 and CPPs-E8) were prepared via non-covalent binding of the peptides and polyanions. Cytotoxicity and anti-proliferative activity were evaluated by MTT assay. Cellular uptake was examined by flow cytometry and fluorescence microscopy. The CPPs exhibited slight cytotoxicity. Binding of polyglutamate to CPPs (CPPs-E4 and CPPs-E8 nanoparticles) decreased their cytotoxicity. CPPs-E8 nanoparticles showed lower cytotoxicity than CPPs-E4 nanoparticles. Cellular uptake of K3W4K3-E8, K2W4K2-E8 and W3K4W3-E8 reached 100% with no difference between each of the mentioned CPPs and its nanoparticles at 50 µM. The anti-proliferative activity of EPR was enhanced following conjugation to peptides and nanoparticles at 25 µM. CPPs-EPR-E4 and CPPs-E8-EPR nanoparticles displayed higher anti-proliferative activity than CPPs-EPR at 25 µM. CPPs-E8-EPR nanoparticles showed higher anti-proliferative activity than CPPs-E4-EPR. K3W4K3-E8-EPR nanoparticles exhibited the highest anti-proliferative activity at 25 µM. The synthesized peptide nanoparticles are proposed as suitable carriers for improving the intracellular delivery of EPR into tumor cells with low cytotoxicity and high antitumor activity.
Collapse
Affiliation(s)
- Samaneh Mohammadi
- a Biotechnology Research Center and Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Parvin Zakeri-Milani
- b Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Nasim Golkar
- c Pharmaceutics Department, School of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Samad Mussa Farkhani
- a Biotechnology Research Center and Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran.,d Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Ali Shirani
- a Biotechnology Research Center and Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran.,d Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Javid Shahbazi Mojarrad
- b Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Ali Nokhodchi
- e Pharmaceutics Research Laboratory, School of Life Sciences , University of Sussex , Brighton , UK
| | - Hadi Valizadeh
- f Drug Applied Research Center and Faculty of Pharmacy , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
26
|
Gong Z, Walls MT, Karley AN, Karlsson AJ. Effect of a Flexible Linker on Recombinant Expression of Cell-Penetrating Peptide Fusion Proteins and Their Translocation into Fungal Cells. Mol Biotechnol 2016; 58:838-849. [DOI: 10.1007/s12033-016-9983-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Caffrey LM, deRonde BM, Minter LM, Tew GN. Mapping Optimal Charge Density and Length of ROMP-Based PTDMs for siRNA Internalization. Biomacromolecules 2016; 17:3205-3212. [PMID: 27599388 PMCID: PMC5094354 DOI: 10.1021/acs.biomac.6b00900] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A fundamental understanding of how polymer structure impacts internalization and delivery of biologically relevant cargoes, particularly small interfering ribonucleic acid (siRNA), is of critical importance to the successful design of improved delivery reagents. Herein we report the use of ring-opening metathesis polymerization (ROMP) methods to synthesize two series of guanidinium-rich protein transduction domain mimics (PTDMs): one based on an imide scaffold that contains one guanidinium moiety per repeat unit, and another based on a diester scaffold that contains two guanidinium moieties per repeat unit. By varying both the degree of polymerization and, in effect, the relative number of cationic charges in each PTDM, the performances of the two ROMP backbones for siRNA internalization were evaluated and compared. Internalization of fluorescently labeled siRNA into Jurkat T cells demonstrated that fluorescein isothiocyanate (FITC)-siRNA internalization had a charge content dependence, with PTDMs containing approximately 40 to 60 cationic charges facilitating the most internalization. Despite this charge content dependence, the imide scaffold yielded much lower viabilities in Jurkat T cells than the corresponding diester PTDMs with similar numbers of cationic charges, suggesting that the diester scaffold is preferred for siRNA internalization and delivery applications. These developments will not only improve our understanding of the structural factors necessary for optimal siRNA internalization, but will also guide the future development of optimized PTDMs for siRNA internalization and delivery.
Collapse
Affiliation(s)
- Leah M Caffrey
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Brittany M deRonde
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Lisa M Minter
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Gregory N Tew
- Department of Polymer Science and Engineering, ‡Department of Veterinary and Animal Sciences, and §Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| |
Collapse
|
28
|
Bera S, Kar RK, Mondal S, Pahan K, Bhunia A. Structural Elucidation of the Cell-Penetrating Penetratin Peptide in Model Membranes at the Atomic Level: Probing Hydrophobic Interactions in the Blood-Brain Barrier. Biochemistry 2016; 55:4982-96. [PMID: 27532224 PMCID: PMC5014585 DOI: 10.1021/acs.biochem.6b00518] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell-penetrating peptides (CPPs) have shown promise in nonpermeable therapeutic drug delivery, because of their ability to transport a variety of cargo molecules across the cell membranes and their noncytotoxicity. Drosophila antennapedia homeodomain-derived CPP penetratin (RQIKIWFQNRRMKWKK), being rich in positively charged residues, has been increasingly used as a potential drug carrier for various purposes. Penetratin can breach the tight endothelial network known as the blood-brain barrier (BBB), permitting treatment of several neurodegenerative maladies, including Alzheimer's disease, Parkinson's disease, and Huntington's disease. However, a detailed structural understanding of penetratin and its mechanism of action is lacking. This study defines structural features of the penetratin-derived peptide, DK17 (DRQIKIWFQNRRMKWKK), in several model membranes and describes a membrane-induced conformational transition of the DK17 peptide in these environments. A series of biophysical experiments, including high-resolution nuclear magnetic resonance spectroscopy, provides the three-dimensional structure of DK17 in different membranes mimicking the BBB or total brain lipid extract. Molecular dynamics simulations support the experimental results showing preferential binding of DK17 to particular lipids at atomic resolution. The peptide conserves the structure of the subdomain spanning residues Ile6-Arg11, despite considerable conformational variation in different membrane models. In vivo data suggest that the wild type, not a mutated sequence, enters the central nervous system. Together, these data highlight important structural and functional attributes of DK17 that could be utilized in drug delivery for neurodegenerative disorders.
Collapse
Affiliation(s)
- Swapna Bera
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| | - Rajiv K Kar
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| | - Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue, Chicago, IL, USA
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| |
Collapse
|
29
|
Abstract
RNA interference mediated gene silencing has tremendous applicability in fields ranging from basic biological research to clinical therapy. However, delivery of siRNA across the cell membrane into the cytoplasm, where the RNA silencing machinery is located, is a significant hurdle in most primary cells. Cell-penetrating peptides (CPPs), peptides that possess an intrinsic ability to translocate across cell membranes, have been explored as a means to achieve cellular delivery of siRNA. Approaches using CPPs by themselves or through incorporation into other siRNA delivery platforms have been investigated with the intent of improving cytoplasmic delivery. Here, we review the utilization of CPPs for siRNA delivery with a focus on strategies developed to enhance cellular uptake, endosomal escape and cytoplasmic localization of CPP/siRNA complexes.
Collapse
|
30
|
Niazi M, Zakeri-Milani P, Najafi Hajivar S, Soleymani Goloujeh M, Ghobakhlou N, Shahbazi Mojarrad J, Valizadeh H. Nano-based strategies to overcome p-glycoprotein-mediated drug resistance. Expert Opin Drug Metab Toxicol 2016; 12:1021-33. [PMID: 27267126 DOI: 10.1080/17425255.2016.1196186] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The discussion about cancer treatment has a long history. Chemotherapy, one of the promising approaches in cancer therapy, is limited in the clinic as plenty of factors evolve and prevent appropriate therapeutic response to drugs. Multi-drug resistance (MDR), which is mostly P-glycoprotein-mediated, is described as the most well-known impediment in this contribution. It extrudes several agents out of cells, arising MDR and decreasing the bioavailability of drugs. Hence, cancer cells become insensitive to chemotherapy. AREAS COVERED Many agents have been developed to reverse MDR, but it is difficult to deliver them into cancer sites and cancer cells. The emerging nano-based drug delivery systems have been more effective to overcome P-glycoprotein-mediated MDR by increasing the intracellular delivery of these agents. Here, we represent systems including siRNA-targeted inhibition of P-gp, monoclonal antibodies, natural extracts, conventional inhibitors, hard nanoparticles and soft nanoparticles as delivery systems in addition to a novel approach applying cell penetrating peptides. EXPERT OPINION Overcoming cancer drug resistance using innovative nanotechnology is being increasingly used and developed. Among resistance mechanisms, drug efflux transporter inhibitors and MDR gene expression silencing are among the those being investigated. In the near future, it seems some of these nanomedical approaches might become the mainstay of effective treatment of important human conditions like cancer.
Collapse
Affiliation(s)
- Mehri Niazi
- a Student Research Committee, Faculty of Advanced Medical Sciences and Research Center for Pharmaceutical Nanotechnology , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Parvin Zakeri-Milani
- b Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Saeedeh Najafi Hajivar
- a Student Research Committee, Faculty of Advanced Medical Sciences and Research Center for Pharmaceutical Nanotechnology , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mehdi Soleymani Goloujeh
- a Student Research Committee, Faculty of Advanced Medical Sciences and Research Center for Pharmaceutical Nanotechnology , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Nasrin Ghobakhlou
- a Student Research Committee, Faculty of Advanced Medical Sciences and Research Center for Pharmaceutical Nanotechnology , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Javid Shahbazi Mojarrad
- c Drug Applied Research Center and Faculty of Pharmacy , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Hadi Valizadeh
- c Drug Applied Research Center and Faculty of Pharmacy , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
31
|
deRonde BM, Posey ND, Otter R, Minter LM, Tew GN. Optimal Hydrophobicity in Ring-Opening Metathesis Polymerization-Based Protein Mimics Required for siRNA Internalization. Biomacromolecules 2016; 17:1969-77. [PMID: 27103189 PMCID: PMC4964964 DOI: 10.1021/acs.biomac.6b00138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Exploring the role of polymer structure for the internalization of biologically relevant cargo, specifically siRNA, is of critical importance to the development of improved delivery reagents. Herein, we report guanidinium-rich protein transduction domain mimics (PTDMs) based on a ring-opening metathesis polymerization scaffold containing tunable hydrophobic moieties that promote siRNA internalization. Structure-activity relationships using Jurkat T cells and HeLa cells were explored to determine how the length of the hydrophobic block and the hydrophobic side chain compositions of these PTDMs impacted siRNA internalization. To explore the hydrophobic block length, two different series of diblock copolymers were synthesized: one series with symmetric block lengths and one with asymmetric block lengths. At similar cationic block lengths, asymmetric and symmetric PTDMs promoted siRNA internalization in the same percentages of the cell population regardless of the hydrophobic block length; however, with 20 repeat units of cationic charge, the asymmetric block length had greater siRNA internalization, highlighting the nontrivial relationships between hydrophobicity and overall cationic charge. To further probe how the hydrophobic side chains impacted siRNA internalization, an additional series of asymmetric PTDMs was synthesized that featured a fixed hydrophobic block length of five repeat units that contained either dimethyl (dMe), methyl phenyl (MePh), or diphenyl (dPh) side chains and varied cationic block lengths. This series was further expanded to incorporate hydrophobic blocks consisting of diethyl (dEt), diisobutyl (diBu), and dicyclohexyl (dCy) based repeat units to better define the hydrophobic window for which our PTDMs had optimal activity. High-performance liquid chromatography retention times quantified the relative hydrophobicities of the noncationic building blocks. PTDMs containing the MePh, diBu, and dPh hydrophobic blocks were shown to have superior siRNA internalization capabilities compared to their more and less hydrophobic counterparts, demonstrating a critical window of relative hydrophobicity for optimal internalization. This better understanding of how hydrophobicity impacts PTDM-induced internalization efficiencies will help guide the development of future delivery reagents.
Collapse
Affiliation(s)
- Brittany M. deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
| | - Nicholas D. Posey
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
| | - Ronja Otter
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| | - Lisa M. Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| | - Gregory N. Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA 01003
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
32
|
Krautwald S, Dewitz C, Fändrich F, Kunzendorf U. Inhibition of regulated cell death by cell-penetrating peptides. Cell Mol Life Sci 2016; 73:2269-84. [PMID: 27048815 PMCID: PMC4887531 DOI: 10.1007/s00018-016-2200-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 12/18/2022]
Abstract
Development of the means to efficiently and continuously renew missing and non-functional proteins in diseased cells remains a major goal in modern molecular medicine. While gene therapy has the potential to achieve this, substantial obstacles must be overcome before clinical application can be considered. A promising alternative approach is the direct delivery of non-permeant active biomolecules, such as oligonucleotides, peptides and proteins, to the affected cells with the purpose of ameliorating an advanced disease process. In addition to receptor-mediated endocytosis, cell-penetrating peptides are widely used as vectors for rapid translocation of conjugated molecules across cell membranes into intracellular compartments and the delivery of these therapeutic molecules is generally referred to as novel prospective protein therapy. As a broad coverage of the enormous amount of published data in this field is unrewarding, this review will provide a brief, focused overview of the technology and a summary of recent studies of the most commonly used protein transduction domains and their potential as therapeutic agents for the treatment of cellular damage and the prevention of regulated cell death.
Collapse
Affiliation(s)
- Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105, Kiel, Germany.
| | - Christin Dewitz
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105, Kiel, Germany
| | - Fred Fändrich
- Clinic for Applied Cellular Medicine, University Hospital Schleswig-Holstein, 24105, Kiel, Germany
| | - Ulrich Kunzendorf
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105, Kiel, Germany
| |
Collapse
|
33
|
Xie X, Lin W, Li M, Yang Y, Deng J, Liu H, Chen Y, Fu X, Liu H, Yang Y. Efficient siRNA Delivery Using Novel Cell-Penetrating Peptide-siRNA Conjugate-Loaded Nanobubbles and Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:1362-1374. [PMID: 27012462 DOI: 10.1016/j.ultrasmedbio.2016.01.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/13/2016] [Accepted: 01/27/2016] [Indexed: 06/05/2023]
Abstract
Because of the absence of tolerable and effective carriers for in vivo delivery, the applications of small interfering RNA (siRNA) in the clinic for therapeutic purposes have been limited. In this study, development of a novel siRNA delivery system based on ultrasound-sensitive nanobubbles (NBs, nano-sized echogenic liposomes) and cell-permeable peptides (CPPs) is described. A CPP-siRNA conjugate was entrapped in an NB, (CPP-siRNA)-NB, and the penetration of CPP-siRNA was temporally masked; local ultrasound stimulation triggered the release of CPP-siRNA from the NBs and activated its penetration. Subsequent research revealed that the (CPP-siRNA)-NBs had a mean particle size of 201 ± 2.05 nm and a siRNA entrapment efficiency >85%. In vitro release results indicated that >90% of the encapsulated CPP-siRNA was released from NBs in the presence of ultrasound, whereas <1.5% (30 min) was released in the absence of ultrasound. Cell experiments indicated higher cellular CPP-siRNA uptake of (CPP-siRNA)-NBs with ultrasound among the various formulations in human breast adenocarcinoma cells (HT-1080). Additionally, after systemic administration in mice, (CPP-siRNA)-NBs accumulated in the tumor, augmented c-myc silencing and delayed tumor progression. In conclusion, the application of (CPP-siRNA)-NBs with ultrasound may constitute an approach to selective targeted delivery of siRNA.
Collapse
Affiliation(s)
- Xiangyang Xie
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Wen Lin
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, China
| | - Mingyuan Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Jianping Deng
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, China
| | - Hui Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Ying Chen
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Xudong Fu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Hong Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Izabela R, Jarosław R, Magdalena A, Piotr R, Ivan K. Transportan 10 improves the anticancer activity of cisplatin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2016. [PMID: 26899863 DOI: 10.1007/s00210-016-1219-5/figures/8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
The aim of this paper was to examine whether cell-penetrating peptides (CPPs) such as transportan 10 (TP10) or protein transduction domain (PTD4) may improve the anticancer activity of cisplatin (cPt). The complexes of TP10 or PTD4 with cPt were used in the experiments. They were carried out on two non-cancer (HEK293 (human embryonic kidney) and HEL299 (human embryo lung)) and two cancer (HeLa (human cervical cancer) and OS143B (human osteosarcoma 143B)) cell lines. Both complexes were tested (MTT assay) with respect to their anticancer or cytotoxic actions. TAMRA (fluorescent dye)-stained preparations were visualized in a fluorescence microscope. The long-term effect of TP10 + cPt and its components on non-cancer and cancer cell lines was observed in inverted phase contrast microscopy. In the MTT test (cell viability assay), the complex of TP10 + cPt produced a more potent effect on the cancer cell lines (HeLa, OS143B) in comparison to that observed after separate treatment with TP10 or cPt. At the same time, the action of the complex and its components was rather small on non-cancer cell lines. On the other hand, a complex of another CPP with cPt, i.e., PTD4 + cPt, was without a significant effect on the cancer cell line (OS143B). The images of the fluorescent microscopy showed TAMRA-TP10 or TAMRA-TP10 + cPt in the interior of the HeLa cells. In the case of TAMRA-PTD4 or TAMRA-PTD4 + cPt, only the first compound was found inside the cancer cell line. In contrast, none of the tested compounds gained access to the interior of the non-cancer cells (HEK293, HEL299). Long-term incubation with the TP10 + cPt (estimated by inverted phase contrast microscopy) lead to an enhanced action of the complex on cell viability (decrease in the number of cells and change in their morphology) as compared with that produced by each single agent. With regard to the tested CPPs, only TP10 improved the anticancer activity of cisplatin if both compounds were used in the form of a complex. Additionally, the complex was relatively safe for non-cancer cells. What is more, TP10 also produced an anticancer effect on HeLa and OS143B cell lines.
Collapse
Affiliation(s)
- Rusiecka Izabela
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | - Rekowski Piotr
- Department of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Kocić Ivan
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
35
|
Backlund CM, Takeuchi T, Futaki S, Tew GN. Relating structure and internalization for ROMP-based protein mimics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:1443-50. [PMID: 27039278 DOI: 10.1016/j.bbamem.2016.03.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/24/2016] [Accepted: 03/29/2016] [Indexed: 12/29/2022]
Abstract
Elucidating the predominant cellular entry mechanism for protein transduction domains (PTDs) and their synthetic mimics (PTDMs) is a complicated problem that continues to be a significant source of debate in the literature. The PTDMs reported here provide a well-controlled platform to vary molecular composition for structure activity relationship studies to further our understanding of PTDs, their non-covalent association with cargo, and their cellular internalization pathways. Specifically, several guanidine rich homopolymers, along with an amphiphilic block copolymer were used to investigate the relationship between structure and internalization activity in HeLa cells, both alone and non-covalently complexed with EGFP by flow cytometery and confocal imaging. The findings indicate that while changing the amount of positive charge on our PTDMs does not seem to affect the endosomal uptake, the presence of hydrophobicity appears to be a critical factor for the polymers to enter cells either alone, or with associated cargo.
Collapse
Affiliation(s)
- Coralie M Backlund
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Toshihide Takeuchi
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Shiroh Futaki
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Gregory N Tew
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
36
|
Büyüktimkin B, Stewart J, Tabanor K, Kiptoo P, Siahaan TJ. Protein and Peptide Conjugates for Targeting Therapeutics and Diagnostics to Specific Cells. Drug Deliv 2016. [DOI: 10.1002/9781118833322.ch20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
37
|
|
38
|
Izabela R, Jarosław R, Magdalena A, Piotr R, Ivan K. Transportan 10 improves the anticancer activity of cisplatin. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:485-97. [PMID: 26899863 PMCID: PMC4823340 DOI: 10.1007/s00210-016-1219-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/08/2016] [Indexed: 11/22/2022]
Abstract
The aim of this paper was to examine whether cell-penetrating peptides (CPPs) such as transportan 10 (TP10) or protein transduction domain (PTD4) may improve the anticancer activity of cisplatin (cPt). The complexes of TP10 or PTD4 with cPt were used in the experiments. They were carried out on two non-cancer (HEK293 (human embryonic kidney) and HEL299 (human embryo lung)) and two cancer (HeLa (human cervical cancer) and OS143B (human osteosarcoma 143B)) cell lines. Both complexes were tested (MTT assay) with respect to their anticancer or cytotoxic actions. TAMRA (fluorescent dye)-stained preparations were visualized in a fluorescence microscope. The long-term effect of TP10 + cPt and its components on non-cancer and cancer cell lines was observed in inverted phase contrast microscopy. In the MTT test (cell viability assay), the complex of TP10 + cPt produced a more potent effect on the cancer cell lines (HeLa, OS143B) in comparison to that observed after separate treatment with TP10 or cPt. At the same time, the action of the complex and its components was rather small on non-cancer cell lines. On the other hand, a complex of another CPP with cPt, i.e., PTD4 + cPt, was without a significant effect on the cancer cell line (OS143B). The images of the fluorescent microscopy showed TAMRA-TP10 or TAMRA-TP10 + cPt in the interior of the HeLa cells. In the case of TAMRA-PTD4 or TAMRA-PTD4 + cPt, only the first compound was found inside the cancer cell line. In contrast, none of the tested compounds gained access to the interior of the non-cancer cells (HEK293, HEL299). Long-term incubation with the TP10 + cPt (estimated by inverted phase contrast microscopy) lead to an enhanced action of the complex on cell viability (decrease in the number of cells and change in their morphology) as compared with that produced by each single agent. With regard to the tested CPPs, only TP10 improved the anticancer activity of cisplatin if both compounds were used in the form of a complex. Additionally, the complex was relatively safe for non-cancer cells. What is more, TP10 also produced an anticancer effect on HeLa and OS143B cell lines.
Collapse
Affiliation(s)
- Rusiecka Izabela
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | - Rekowski Piotr
- Department of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Kocić Ivan
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
39
|
Gao D, Lin XP, Zhang ZP, Li W, Men D, Zhang XE, Cui ZQ. Intracellular cargo delivery by virus capsid protein-based vehicles: From nano to micro. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 12:365-76. [PMID: 26711962 DOI: 10.1016/j.nano.2015.10.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/27/2015] [Accepted: 10/29/2015] [Indexed: 12/31/2022]
Abstract
UNLABELLED Cellular delivery is an important concern for the efficiency of medicines and sensors for disease diagnoses and therapy. However, this task is quite challenging. Self-assembly virus capsid proteins might be developed as building blocks for multifunctional cellular delivery vehicles. In this work, we found that SV40 VP1 (Simian virus 40 major capsid protein) could function as a new cell-penetrating protein. The VP1 protein could carry foreign proteins into cells in a pentameric structure. A double color structure, with red QDs (Quantum dots) encapsulated by viral capsids fused with EGFP, was created for imaging cargo delivery and release from viral capsids. The viral capsids encapsulating QDs were further used for cellular delivery of micron-sized iron oxide particles (MPIOs). MPIOs were efficiently delivered into live cells and controlled by a magnetic field. Therefore, our study built virus-based cellular delivery systems for different sizes of cargos: protein molecules, nanoparticles, and micron-sized particles. FROM THE CLINICAL EDITOR Much research is being done to investigate methods for efficient and specific cellular delivery of drugs, proteins or genetic material. In this article, the authors describe their approach in using self-assembly virus capsid proteins SV40 VP1 (Simian virus 40 major capsid protein). The cell-penetrating behavior provided excellent cellular delivery and should give a new method for biomedical applications.
Collapse
Affiliation(s)
- Ding Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Xiu-Ping Lin
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Zhi-Ping Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Dong Men
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xian-En Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China; National Key Laboratory of Macrobiomolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zong-Qiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
40
|
Saghatelian A, Couso JP. Discovery and characterization of smORF-encoded bioactive polypeptides. Nat Chem Biol 2015; 11:909-16. [PMID: 26575237 PMCID: PMC4956473 DOI: 10.1038/nchembio.1964] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 10/19/2015] [Indexed: 12/13/2022]
Abstract
Analysis of genomes, transcriptomes and proteomes reveals the existence of hundreds to thousands of translated, yet non-annotated, short open reading frames (small ORFs or smORFs). The discovery of smORFs and their protein products, smORF-encoded polypeptides (SEPs), points to a fundamental gap in our knowledge of protein-coding genes. Various studies have identified central roles for smORFs in metabolism, apoptosis and development. The discovery of these bioactive SEPs emphasizes the functional potential of this unexplored class of biomolecules. Here, we provide an overview of this emerging field and highlight the opportunities for chemical biology to answer fundamental questions about these novel genes. Such studies will provide new insights into the protein-coding potential of genomes and identify functional genes with roles in biology and disease.
Collapse
Affiliation(s)
- Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, San Diego, CA 92037
| | - Juan Pablo Couso
- School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 6PU, UK
| |
Collapse
|
41
|
Xie X, Yang Y, Lin W, Liu H, Liu H, Yang Y, Chen Y, Fu X, Deng J. Cell-penetrating peptide-siRNA conjugate loaded YSA-modified nanobubbles for ultrasound triggered siRNA delivery. Colloids Surf B Biointerfaces 2015; 136:641-50. [PMID: 26492155 DOI: 10.1016/j.colsurfb.2015.10.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 09/29/2015] [Accepted: 10/03/2015] [Indexed: 10/22/2022]
Abstract
Due to the absence of effective in vivo delivery systems, the employment of small interference RNA (siRNA) in the clinic has been hindered. In this paper, a new siRNA targeting system for EphA2-positive tumors was developed, based on ultrasound-sensitive nanobubbles (NBs) and cell-permeable peptides (CPPs). Here, a CPP-siRNA conjugate (CPP-siRNA) was entrapped in an ephrin mimetic peptide (YSA peptide)-modified NB (CPP-siRNA/YSA-NB) and the penetration of the CPP-siRNA was temporally masked; local ultrasound stimulation triggered the release of CPP-siRNA from the NBs and activated its penetration. Subsequent research demonstrated that the CPP-siRNA/YSA-NBs had particle sizes of approximately 200 nm and a siRNA entrapment efficiency of more than 85%. The in vitro release results showed that over 90% of the encapsulated CPP-siRNA released from the NBs in the presence of ultrasound, while less than 1.5% of that (30 min) released without ultrasound. Cell experiments showed a the higher CPP-siRNA cellular uptake of CPP-siRNA/YSA-NB among the various formulations in human breast adenocarcinoma cells (MCF-7, EphA2 positive cells). Additionally, after systemic administration in mice, CPP-siRNA/YSA-NB accumulated in the tumor, augmented c-Myc silencing and delayed tumor progression. In conclusion, the application of CPP-siRNA/YSA-NB with ultrasound may provide a strategy for the selective and efficient delivery of siRNA.
Collapse
Affiliation(s)
- Xiangyang Xie
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan 430070, PR China
| | - Yanfang Yang
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Wen Lin
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi 435000, PR China
| | - Hui Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan 430070, PR China
| | - Hong Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan 430070, PR China
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, No. 27 Taiping Road, Beijing 100850, PR China.
| | - Ying Chen
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan 430070, PR China
| | - Xudong Fu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan 430070, PR China
| | - Jianping Deng
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi 435000, PR China.
| |
Collapse
|
42
|
deRonde BM, Torres JA, Minter LM, Tew GN. Development of Guanidinium-Rich Protein Mimics for Efficient siRNA Delivery into Human T Cells. Biomacromolecules 2015; 16:3172-9. [PMID: 26324222 DOI: 10.1021/acs.biomac.5b00795] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RNA interference is gaining attention as a means to explore new molecular pathways and for its potential as a therapeutic; however, its application in immortal and primary T cells is limited due to challenges with efficient delivery in these cell types. Herein, we report the development of guanidinium-rich protein transduction domain mimics (PTDMs) based on a ring-opening metathesis polymerization scaffold that delivers siRNA into Jurkat T cells and human peripheral blood mononuclear cells (hPBMCs). Homopolymer and block copolymer PTDMs with varying numbers of guanidinium moieties were designed and tested to assess the effect cationic charge content and the addition of a segregated, hydrophobic block had on siRNA internalization and delivery. Internalization of fluorescently labeled siRNA into Jurkat T cells illustrates that the optimal cationic charge content, 40 charges per polymer, leads to higher efficiencies, with block copolymers outperforming their homopolymer counterparts. PTDMs also outperformed commercial reagents commonly used for siRNA delivery applications. Select PTDM candidates were further screened to assess the role the PTDM structure has on the delivery of biologically active siRNA into primary cells. Specifically, siRNA to hNOTCH1 was delivered to hPBMCs enabling 50-80% knockdown efficiencies, with longer PTDMs showing improved protein reduction. By evaluating the PTDM design parameters for siRNA delivery, more efficient PTDMs were discovered that improved delivery and gene (NOTCH) knockdown in T cells. Given the robust delivery of siRNA by these novel PTDMs, their development should aid in the exploration of T cell molecular pathways leading eventually to new therapeutics.
Collapse
Affiliation(s)
- Brittany M deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Joe A Torres
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Lisa M Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| | - Gregory N Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Department of Veterinary and Animal Sciences, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst , Amherst, Massachusetts 01003, United States
| |
Collapse
|
43
|
Bilichak A, Luu J, Eudes F. Intracellular delivery of fluorescent protein into viable wheat microspores using cationic peptides. FRONTIERS IN PLANT SCIENCE 2015; 6:666. [PMID: 26379691 PMCID: PMC4552043 DOI: 10.3389/fpls.2015.00666] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 08/13/2015] [Indexed: 05/24/2023]
Abstract
Microspores are specialized generative cells with haploid genome that demonstrate the amenability toward embryogenesis under certain conditions. The induced microspore culture technique is largely exploited by the breeding programs of wheat and other crops due to its high efficiency for generation of the large number of haploid plants in the relatively short period of time. The ability to produce mature double haploid plant from a single cell has also attracted attention of the plant biotechnologists in the past few years. More importantly, the possibility to deliver proteins for improvement of embryogenesis and the genome modification purposes holds great potential for transgene-free wheat biotechnology. In the present study, we examined the ability of cationic and amphipathic cell penetrating peptides (CPPs) to convey a covalently-linked mCherry protein inside the viable microspores. We demonstrate that the affinity of CPPs to the microspore cells dependents on their charge with the highest efficiency of CPP-mCherry binding to the cells achieved by cationic CPPs (penetratin and R9). Additionally, due to overall negative charge of the microspore cell wall, the successful uptake of the protein cargo by live microspore cells is attained by utilization of a reversible disulfide bond between the R9 CPP and mCherry protein. Overall, the approach proposed herein can be applied by the other biotechnology groups for the fast and efficient screening of the different CPP candidates for their ability to deliver proteins inside the viable plant cells.
Collapse
Affiliation(s)
- Andriy Bilichak
- Lethbridge Research Centre, Agriculture and Agri-Food CanadaLethbridge, AB, Canada
| | | | | |
Collapse
|
44
|
Lehto T, Wagner E. Sequence-defined polymers for the delivery of oligonucleotides. Nanomedicine (Lond) 2015; 9:2843-59. [PMID: 25535686 DOI: 10.2217/nnm.14.166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Short synthetic oligonucleotides (ONs) are a group of therapeutic molecules with enormous clinical potential owing to their high specificity and ability to target the expression of virtually any single or group of genes. Clinical translation of ONs is hampered by the inadequate bioavailability in the target cells due to the substantial extracellular and intracellular barriers exposed to these molecules. Different cationic polymers have been successfully deployed for the delivery of ONs. However, heterogeneous nature of these classical polymers is not suitable for clinical applications and hence vectors with completely defined structure are required. In this review, we discuss recent advances with sequence-defined polymers and their application for the delivery of short ONs.
Collapse
Affiliation(s)
- Taavi Lehto
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Munich, Germany
| | | |
Collapse
|
45
|
Xie X, Lin W, Liu H, Deng J, Chen Y, Liu H, Fu X, Yang Y. Ultrasound-responsive nanobubbles contained with peptide–camptothecin conjugates for targeted drug delivery. Drug Deliv 2015; 23:2756-2764. [PMID: 26289216 DOI: 10.3109/10717544.2015.1077289] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Xiangyang Xie
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan, PR China
| | - Wen Lin
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, PR China, and
| | - Hui Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan, PR China
| | - Jianping Deng
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, PR China, and
| | - Ying Chen
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan, PR China
| | - Hong Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan, PR China
| | - Xudong Fu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Command, Wuhan, PR China
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| |
Collapse
|
46
|
Lin W, Xie X, Deng J, Liu H, Chen Y, Fu X, Liu H, Yang Y. Cell-penetrating peptide-doxorubicin conjugate loaded NGR-modified nanobubbles for ultrasound triggered drug delivery. J Drug Target 2015; 24:134-46. [PMID: 26176270 DOI: 10.3109/1061186x.2015.1058802] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A new drug-targeting system for CD13(+) tumors has been developed, based on ultrasound-sensitive nanobubbles (NBs) and cell-permeable peptides (CPPs). Here, the CPP-doxorubicin conjugate (CPP-DOX) was entrapped in the asparagine-glycine-arginine (NGR) peptide modified NB (CPP-DOX/NGR-NB) and the penetration of CPP-DOX was temporally masked; local ultrasound stimulation could trigger the CPP-DOX release from NB and activate its penetration. The CPP-DOX/NGR-NBs had particle sizes of about 200 nm and drug entrapment efficiency larger than 90%. In vitro release results showed that over 85% of the encapsulated DOX or CPP-DOX would release from NBs in the presence of ultrasound, while less than 1.5% of that (30 min) without ultrasound. Cell experiments showed the higher cellular CPP-DOX uptake of CPP-DOX/NGR-NB among the various NB formulations in Human fibrosarcoma cells (HT-1080, CD13(+)). The CPP-DOX/NGR-NB with ultrasound treatment exhibited an increased cytotoxic activity than the one without ultrasound. In nude mice xenograft of HT-1080 cells, CPP-DOX/NGR-NB with ultrasound showed a higher tumor inhibition effect (3.1% of T/C%, day 24), longer median survival time (50 days) and excellent body safety compared with the normal DOX injection group. These results indicate that the constructed vesicle would be a promising drug delivery system for specific cancer treatment.
Collapse
Affiliation(s)
- Wen Lin
- a Department of Clinical Laboratory , Huangshi Love & Health Hospital of Hubei Province , Huangshi , People's Republic of China
| | - Xiangyang Xie
- b Department of Pharmacy , Wuhan General Hospital of Guangzhou Military Command , Wuhan , People's Republic of China , and
| | - Jianping Deng
- a Department of Clinical Laboratory , Huangshi Love & Health Hospital of Hubei Province , Huangshi , People's Republic of China
| | - Hui Liu
- b Department of Pharmacy , Wuhan General Hospital of Guangzhou Military Command , Wuhan , People's Republic of China , and
| | - Ying Chen
- b Department of Pharmacy , Wuhan General Hospital of Guangzhou Military Command , Wuhan , People's Republic of China , and
| | - Xudong Fu
- b Department of Pharmacy , Wuhan General Hospital of Guangzhou Military Command , Wuhan , People's Republic of China , and
| | - Hong Liu
- b Department of Pharmacy , Wuhan General Hospital of Guangzhou Military Command , Wuhan , People's Republic of China , and
| | - Yang Yang
- c Department of Pharmacy , Beijing Institute of Pharmacology and Toxicology , Beijing , People's Republic of China
| |
Collapse
|
47
|
deRonde BM, Tew GN. Development of protein mimics for intracellular delivery. Biopolymers 2015; 104:265-80. [PMID: 25858701 PMCID: PMC4516575 DOI: 10.1002/bip.22658] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/24/2015] [Accepted: 03/30/2015] [Indexed: 12/19/2022]
Abstract
Designing delivery agents for therapeutics is an ongoing challenge. As treatments and desired cargoes become more complex, the need for improved delivery vehicles becomes critical. Excellent delivery vehicles must ensure the stability of the cargo, maintain the cargo's solubility, and promote efficient delivery and release. In order to address these issues, many research groups have looked to nature for design inspiration. Proteins, such as HIV-1 trans-activator of transcription (TAT) and Antennapedia homeodomain protein, are capable of crossing cellular membranes. However, due to the complexities of their structures, they are synthetically challenging to reproduce in the laboratory setting. Being able to incorporate the key features of these proteins that enable cell entry into simpler scaffolds opens up a wide range of opportunities for the development of new delivery reagents with improved performance. This review charts the development of protein mimics based on cell-penetrating peptides (CPPs) and how structure-activity relationships (SARs) with these molecules and their protein counterparts ultimately led to the use of polymeric scaffolds. These scaffolds deviate from the normal peptide backbone, allowing for simpler, synthetic procedures to make carriers and tune chemical compositions for application specific needs. Successful design of polymeric protein mimics would allow researchers to further understand the key features in proteins and peptides necessary for efficient delivery and to design the next generation of more efficient delivery reagents.
Collapse
Affiliation(s)
- Brittany M deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003
| | - Gregory N Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, 01003
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA, 01003
| |
Collapse
|
48
|
Ming X, Laing B. Bioconjugates for targeted delivery of therapeutic oligonucleotides. Adv Drug Deliv Rev 2015; 87:81-9. [PMID: 25689735 DOI: 10.1016/j.addr.2015.02.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 01/05/2023]
Abstract
Bioconjugates have been used to deliver therapeutic oligonucleotides to their pharmacological targets in diseased cells. Molecular-scale conjugates can be prepared by directly linking targeting ligands with oligonucleotides and the resultant conjugates can selectively bind to cell surface receptors in target cells in diseased tissues. Besides targeted delivery, additional functionality can be incorporated in the conjugates by utilization of carrier molecules, and these larger conjugates are called carrier-associated conjugates. Both molecular and carrier-associated conjugates have achieved initial successes in clinical trials for treating liver diseases; therefore, currently the greater challenge is to deliver oligonucleotides to extrahepatic tissues such as tumors. This review will provide an update on the application of oligonucleotide conjugates for targeted delivery during the last decade. By identifying key elements for successful delivery, it is suggested that oligonucleotide conjugates with intermediate size, cell targeting ability, and endosomal release functionality are superior systems to advance oligonucleotides to achieve their full therapeutic potentials.
Collapse
Affiliation(s)
- Xin Ming
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Brian Laing
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
49
|
Lönn P, Dowdy SF. Cationic PTD/CPP-mediated macromolecular delivery: charging into the cell. Expert Opin Drug Deliv 2015; 12:1627-36. [PMID: 25994800 DOI: 10.1517/17425247.2015.1046431] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Macromolecular therapeutics, including enzymes, transcription factors, siRNAs, peptides and large synthetic molecules, can potentially be used to treat human diseases by targeting intracellular molecular pathways and modulating biological responses. However, large macromolecules have no ability to enter cells and require delivery vehicles. Protein transduction domains (PTDs), also known as cell-penetrating peptides (CPPs), are a diverse class of peptides that can deliver macromolecules into cells. AREAS COVERED In this review, we cover the uptake and usage of arginine-rich PTDs/CPPs (TAT-PTD, Penetratin/Antp and 8R). We review the endocytosis-mediated uptake of these peptides and highlight three important steps: i) cell association; ii) internalization and iii) endosomal escape. We also discuss the array of different cargos that have been delivered by cationic PTDs/CPPs as well as cellular processes and biological responses that have been modulated. EXPERT OPINION PTDs/CPPs have shown great potential to deliver otherwise undeliverable macromolecular therapeutics into cells for experimentation in cell culture and in animal disease models in vivo. Moreover, over 25 clinical trials have been performed predominantly using the TAT-PTD. However, more work is still needed. Endosomal escape and target-cell specificity remain two of the major future challenges.
Collapse
Affiliation(s)
- Peter Lönn
- a 1 UCSD School of Medicine, Department of Cellular and Molecular Medicine , 9500 Gilman Dr., La Jolla, CA 92093-0686, USA .,b 2 Uppsala University, Science for Life Laboratory, Department of Immunology, Genetics and Pathology , SE-751 08 Uppsala, Sweden
| | - Steven F Dowdy
- a 1 UCSD School of Medicine, Department of Cellular and Molecular Medicine , 9500 Gilman Dr., La Jolla, CA 92093-0686, USA
| |
Collapse
|
50
|
Durzyńska J, Przysiecka Ł, Nawrot R, Barylski J, Nowicki G, Warowicka A, Musidlak O, Goździcka-Józefiak A. Viral and Other Cell-Penetrating Peptides as Vectors of Therapeutic Agents in Medicine. J Pharmacol Exp Ther 2015; 354:32-42. [DOI: 10.1124/jpet.115.223305] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022] Open
|