1
|
Wang Y, Zou R, Zhou Y, Zheng Y, Peng C, Liu Y, Tan H, Fu Q, Ding M. Unraveling mechanisms of protein encapsulation and release in coacervates via molecular dynamics and machine learning. Chem Sci 2024; 15:13442-13451. [PMID: 39183928 PMCID: PMC11339950 DOI: 10.1039/d4sc03061c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Coacervates play a pivotal role in protein-based drug delivery research, yet their drug encapsulation and release mechanisms remain poorly understood. Here, we utilized the Martini model to investigate bovine serum albumin (BSA) protein encapsulation and release within polylysine/polyglutamate (PLys/PGlu) coacervates. Our findings emphasize the importance of ingredient addition sequence in coacervate formation and encapsulation rates, attributed to preference contact between oppositely charged proteins and poly(amino acid)s. Notably, coacervates composed of β-sheet poly(amino acid)s demonstrate greater BSA encapsulation efficiency due to their reduced entropy and flexibility. Furthermore, we examined the pH responsiveness of coacervates, shedding light on the dissolution process driven by Coulomb forces. By leveraging machine learning algorithms to analyze simulation results, our research advances the understanding of coacervate-based drug delivery systems, with the ultimate goal of optimizing therapeutic outcomes.
Collapse
Affiliation(s)
- Yiwei Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Rongrong Zou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Yeqiang Zhou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Yi Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Chuan Peng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Yang Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Qiang Fu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| |
Collapse
|
2
|
Hirano M, Yokoo H, Goto C, Oba M, Misawa T, Demizu Y. Magainin 2-derived stapled peptides derived with the ability to deliver pDNA, mRNA, and siRNA into cells. Chem Sci 2023; 14:10403-10410. [PMID: 37799999 PMCID: PMC10548513 DOI: 10.1039/d3sc04124g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/27/2023] [Indexed: 10/07/2023] Open
Abstract
We have developed cell-penetrating stapled peptides based on the amphipathic antimicrobial peptide magainin 2 for intracellular delivery of nucleic acids such as pDNA, mRNA, and siRNA. Various types of stapled peptides with a cross-linked structure were synthesised in the hydrophobic region of the amphipathic structure, and their efficacy in intracellular delivery of pDNA was evaluated. The results showed that the stapled peptide st7-5 could deliver pDNA into cells. To improve the deliverability of st7-5, we further designed st7-5_R, in which the Lys residues were replaced by Arg residues. The peptide st7-5_R formed compact and stable complexes with pDNA and was able to efficiently transfer pDNA into the cell. In addition to pDNA, st7-5_R was also able to deliver mRNA and siRNA into the cell. Thus, st7-5_R is a novel peptide that can achieve efficient intracellular delivery of three different nucleic acids.
Collapse
Affiliation(s)
- Motoharu Hirano
- Division of Organic Chemistry, National Institute of Health Sciences 3-25-26 Tonomachi Kawasaki Kanagawa 210-9501 Japan
- Graduate School of Medical Life Science, Yokohama City University 1-7-29 Yokohama Kanagawa 230-0045 Japan
| | - Hidetomo Yokoo
- Division of Organic Chemistry, National Institute of Health Sciences 3-25-26 Tonomachi Kawasaki Kanagawa 210-9501 Japan
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kyoto 606-0823 Japan
| | - Chihiro Goto
- Division of Organic Chemistry, National Institute of Health Sciences 3-25-26 Tonomachi Kawasaki Kanagawa 210-9501 Japan
- Graduate School of Medical Life Science, Yokohama City University 1-7-29 Yokohama Kanagawa 230-0045 Japan
| | - Makoto Oba
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine Kyoto 606-0823 Japan
| | - Takashi Misawa
- Division of Organic Chemistry, National Institute of Health Sciences 3-25-26 Tonomachi Kawasaki Kanagawa 210-9501 Japan
| | - Yosuke Demizu
- Division of Organic Chemistry, National Institute of Health Sciences 3-25-26 Tonomachi Kawasaki Kanagawa 210-9501 Japan
- Graduate School of Medical Life Science, Yokohama City University 1-7-29 Yokohama Kanagawa 230-0045 Japan
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Division of Pharmaceutical Science of Okayama University 1-1-1 Tsushimanaka Kita 700-8530 Japan
| |
Collapse
|
3
|
Luo W, Homma C, Hayamizu Y. Rational Design and Self-Assembly of Histidine-Rich Peptides on a Graphite Surface. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:7057-7062. [PMID: 37171391 DOI: 10.1021/acs.langmuir.3c00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Histidine-rich peptides (HRPs) have been investigated to create functional biomolecules based on the nature of histidine, such as ion binding and catalytic activity. The organization of these HRPs on a solid surface can lead to surface functionalization with the well-known properties of HRPs. However, immobilization of HRPs on the surface has not been realized. Here, we design a series of octapeptides with histidine repeat units, aiming to establish their self-assembly on a graphite surface to produce a highly robust and active nanoscaffold. The new design has (XH)4, and we incorporated various types of hydrophobic amino acids at X in the sequence to facilitate their interaction with the surface. The effect of the pair of amino acids on their self-assembly was investigated by atomic force microscopy. Contact angle measurement revealed that these assemblies functionalized graphite surfaces with different wetting chemistry. Moreover, the secondary structure of peptides was characterized by Fourier transform infrared spectroscopy (FTIR), which gives us further insights into the conformation of histidine repeat peptides on the surface. Our results showed a new approach to applying histidine-rich peptides on the surface and tuning the self-assembly behavior by introducing different counter amino acids that could be integrated with a wide range of biosensing and biotechnology applications.
Collapse
Affiliation(s)
- Wei Luo
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguroku, Tokyo 152-8550, Japan
| | - Chishu Homma
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguroku, Tokyo 152-8550, Japan
| | - Yuhei Hayamizu
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguroku, Tokyo 152-8550, Japan
| |
Collapse
|
4
|
Zhong W, Huang L, Lin Y, Xing C, Lu C. Endogenous dual miRNA-triggered dynamic assembly of DNA nanostructures for in-situ dual siRNA delivery. SCIENCE CHINA MATERIALS 2023; 66:1-9. [PMID: 37362200 PMCID: PMC10163297 DOI: 10.1007/s40843-022-2420-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/06/2023] [Indexed: 06/28/2023]
Abstract
A theranostic strategy of multiple microRNA (miRNA)-triggered in-situ delivery of small interfering RNA (siRNA) can effectively improve the precise therapy of cancer cells. Benefiting from the advantages of programmability, specific molecular recognition, easy functionalization and marked biocompatibility of DNA nanostructures, we designed a three-dimensional (3D) DNA nano-therapeutic platform for dual miRNA-triggered in-situ delivery of siRNA. The 3D DNA nanostructure (TY1Y2) was constructed based on the self-assembly of a DNA tetrahedra scaffold, two sets of Y-shaped DNA (Y1 and Y2), and EpCAM-aptamer which functionalized as the ligand molecule for the recognition of specific cancer cells. After being specifically internalized into the targeted cancer cells, TY1Y2 was triggered by two endogenous miRNAs (miR-21 and miR-122), resulting in the generation of strong fluorescence resonance energy transfer fluorescent signal for dual miRNAs imaging. Meanwhile, the therapeutic siRNAs (siSurvivin and siBcl2) could also be in-situ generated and released from TY1Y2 through the strand-displacement reactions for the synergistic gene therapy of cancer cells. This 3D DNA nanostructure integrated the specific imaging of endogenous biomarkers and the in-situ delivery of therapeutic genes into the multifunctional nanoplatform, revealing the promising applications for the diagnosis and treatment of cancer. Electronic Supplementary Material Supplementary material is available in the online version of this article at 10.1007/s40843-022-2420-y.
Collapse
Affiliation(s)
- Wukun Zhong
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116 China
| | - Lei Huang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116 China
| | - Yuhong Lin
- Institute of Nanobiomaterials and Immunology, School of Life Science, Taizhou University, Taizhou, 318000 China
| | - Chao Xing
- Fujian Key Laboratory of Functional Marine Sensing Materials, Center for Advanced Marine Materials and Smart Sensors, Minjiang University, Fuzhou, 350108 China
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350116 China
| |
Collapse
|
5
|
Elizarova TN, Antopolsky ML, Novichikhin DO, Skirda AM, Orlov AV, Bragina VA, Nikitin PI. A Straightforward Method for the Development of Positively Charged Gold Nanoparticle-Based Vectors for Effective siRNA Delivery. Molecules 2023; 28:molecules28083318. [PMID: 37110552 PMCID: PMC10144622 DOI: 10.3390/molecules28083318] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The therapeutic potential of short interfering RNA (siRNA) to treat many diseases that are incurable with traditional preparations is limited by the extensive metabolism of serum nucleases, low permeability through biological membrane barriers because of a negative charge, and endosomal trapping. Effective delivery vectors are required to overcome these challenges without causing unwanted side effects. Here, we present a relatively simple synthetic protocol to obtain positively charged gold nanoparticles (AuNPs) with narrow size distribution and the surface modified with Tat-related cell-penetrating peptide. The AuNPs were characterized using TEM and the localized surface plasmon resonance technique. The synthesized AuNPs showed low toxicity in experiments in vitro and were able to effectively form complexes with double-stranded siRNA. The obtained delivery vehicles were used for intracellular delivery of siRNA in an ARPE-19 cell line transfected with secreted embryonic alkaline phosphatase (SEAP). The delivered oligonucleotide remained intact and caused a significant knockdown effect on SEAP cell production. The developed material could be useful for delivery of negatively charged macromolecules, such as antisense oligonucleotides and various RNAs, particularly for retinal pigment epithelial cell drug delivery.
Collapse
Affiliation(s)
- Tatiana N Elizarova
- Prokhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Maxim L Antopolsky
- Prokhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Denis O Novichikhin
- Prokhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russia
| | - Artemiy M Skirda
- Prokhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russia
| | - Alexey V Orlov
- Prokhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vera A Bragina
- Prokhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Petr I Nikitin
- Prokhorov General Physics Institute, Russian Academy of Sciences, 119991 Moscow, Russia
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russia
| |
Collapse
|
6
|
Ma H, Cao M. Designed Peptide Assemblies for Efficient Gene Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:13627-13634. [PMID: 36318179 DOI: 10.1021/acs.langmuir.2c02197] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The safe and efficient delivery of nucleic acids including DNA, mRNA, siRNA, and miRNA into targeted cells is critical for gene therapy. Currently, viral gene vectors are very popular, but they have potential toxicity and insecurity. Therefore, the development of nonviral vectors has attracted considerable research attention. Peptide assemblies are superior candidates for being used as gene vectors by having good biocompatibility, versatile molecular design, excellent assembly capacity, ease of modification, and stimuli responsivity. The de novo designed peptides not only can induce efficient condensation of nucleic acids into compacted nanoparticles and protect them from enzymatic digestion but also can effectively overcome biological barriers and improve gene delivery efficiency through targeted delivery, enhanced cellular uptake, improved endolysosomal escape, and nuclear importation. By having these merits, peptidic gene vectors are developing fast, showing outstanding advantages compared to liposome and polymer vectors. This Perspective focuses on peptidic gene delivery systems by emphasizing the molecular design strategies for meeting the criteria of gene condensation, protection from nuclease degradation, cellular uptake, endolysosomal escape, and so on. The new arising research area of peptide-based artificial viruses for gene and ribonucleoprotein delivery has also been reviewed. The challenges and future perspectives are put forward, aiming to provide a conclusive guide for the development of peptidic delivery systems to achieve efficient gene therapy.
Collapse
Affiliation(s)
- Hongchao Ma
- State Key Laboratory of Heavy Oil Processing and Department of Biological and Energy Chemical Engineering, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Meiwen Cao
- State Key Laboratory of Heavy Oil Processing and Department of Biological and Energy Chemical Engineering, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
7
|
Adak A, Das G, Gupta V, Khan J, Mukherjee N, Mondal P, Roy R, Barman S, Gharai PK, Ghosh S. Evolution of Potential Antimitotic Stapled Peptides from Multiple Helical Peptide Stretches of the Tubulin Heterodimer Interface: Helix-Mimicking Stapled Peptide Tubulin Inhibitors. J Med Chem 2022; 65:13866-13878. [PMID: 36240440 DOI: 10.1021/acs.jmedchem.2c01116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein-protein interactions play a crucial role in microtubule dynamics. Microtubules are considered as a key target for the design and development of anticancer therapeutics, where inhibition of tubulin-tubulin interactions plays a crucial role. Here, we focused on a few key helical stretches at the interface of α,β-tubulin heterodimers and developed a structural mimic of these helical peptides, which can serve as potent inhibitors of microtubule polymerization. To induce helicity, we have made stapled analogues of these sequences. Thereafter, we modified the lead sequences of the antimitotic stapled peptides with halo derivatives. It is observed that halo-substituted stapled peptides follow an interesting trend for the electronegativity of halogen atoms in interaction patterns with tubulin and a correlation in the toxicity profile. Remarkably, we found that para-fluorophenylalanine-modified stapled peptide is the most potent inhibitors, which perturbs microtubule dynamics, induces apoptotic death, and inhibits the growth of melanoma.
Collapse
Affiliation(s)
- Anindyasundar Adak
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India
| | - Gaurav Das
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India
| | - Varsha Gupta
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India
| | - Juhee Khan
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India
| | - Nabanita Mukherjee
- Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology Jodhpur, NH 62, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Prasenjit Mondal
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India
| | - Rajsekhar Roy
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Surajit Barman
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India
| | - Prabir Kumar Gharai
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India
| | - Surajit Ghosh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal 700 032, India.,Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India.,Smart Healthcare Department, Interdisciplinary Research Platform, Indian Institute of Technology Jodhpur, NH 62, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| |
Collapse
|
8
|
Zhao B, Chen S, Hong Y, Jia L, Zhou Y, He X, Wang Y, Tian Z, Yang Z, Gao D. Research Progress of Conjugated Nanomedicine for Cancer Treatment. Pharmaceutics 2022; 14:1522. [PMID: 35890416 PMCID: PMC9315807 DOI: 10.3390/pharmaceutics14071522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 12/05/2022] Open
Abstract
The conventional cancer therapeutic modalities include surgery, chemotherapy and radiotherapy. Although immunotherapy and targeted therapy are also widely used in cancer treatment, chemotherapy remains the cornerstone of tumor treatment. With the rapid development of nanotechnology, nanomedicine is believed to be an emerging field to further improve the efficacy of chemotherapy. Until now, there are more than 17 kinds of nanomedicine for cancer therapy approved globally. Thereinto, conjugated nanomedicine, as an important type of nanomedicine, can not only possess the targeted delivery of chemotherapeutics with great precision but also achieve controlled drug release to avoid adverse effects. Meanwhile, conjugated nanomedicine provides the platform for combining several different therapeutic approaches (chemotherapy, photothermal therapy, photodynamic therapy, thermodynamic therapy, immunotherapy, etc.) with the purpose of achieving synergistic effects during cancer treatment. Therefore, this review focuses on conjugated nanomedicine and its various applications in synergistic chemotherapy. Additionally, the further perspectives and challenges of the conjugated nanomedicine are also addressed, which clarifies the design direction of a new generation of conjugated nanomedicine and facilitates the translation of them from the bench to the bedside.
Collapse
Affiliation(s)
- Bin Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
- Department of Epidemiology, Shaanxi Provincial Cancer Hospital, Xi’an 710061, China
| | - Sa Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
- Shaanxi Provincial Centre for Disease Control and Prevention, Xi’an 710054, China
| | - Ye Hong
- Center of Digestive Endoscopy, Shaanxi Provincial Cancer Hospital, Xi’an 710061, China;
| | - Liangliang Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
| | - Ying Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
| | - Xinyu He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
| | - Ying Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
- Research Institute of Xi’an Jiaotong University, Hangzhou 311200, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (B.Z.); (S.C.); (L.J.); (Y.Z.); (X.H.); (Y.W.); (Z.T.)
| |
Collapse
|
9
|
Abstract
AbstractBiophysical studies have a very high impact on the understanding of internalization, molecular mechanisms, interactions, and localization of CPPs and CPP/cargo conjugates in live cells or in vivo. Biophysical studies are often first carried out in test-tube set-ups or in vitro, leading to the complicated in vivo systems. This review describes recent studies of CPP internalization, mechanisms, and localization. The multiple methods in these studies reveal different novel and important aspects and define the rules for CPP mechanisms, hopefully leading to their improved applicability to novel and safe therapies.
Collapse
Affiliation(s)
- Matjaž Zorko
- University of Ljubljana, Medical Faculty, Institute of Biochemistry and Molecular Genetics, Vrazov trg 2, 1000Ljubljana, Slovenia,
| | - Ülo Langel
- University of Stockholm, Department of Biochemistry and Biophysics, Svante Arrhenius väg 16, 106 91 Stockholm, Sweden, , and Institute of Technology, University of Tartu, Nooruse 1, Tartu, Estonia, 50411
| |
Collapse
|
10
|
Wang J, Chen G, Liu N, Han X, Zhao F, Zhang L, Chen P. Strategies for improving the safety and RNAi efficacy of noncovalent peptide/siRNA nanocomplexes. Adv Colloid Interface Sci 2022; 302:102638. [PMID: 35299136 DOI: 10.1016/j.cis.2022.102638] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 12/12/2022]
Abstract
In the past decades, the striking development of cationic polypeptides and cell-penetrating peptides (CPPs) tailored for small interfering RNA (siRNA) delivery has been fuelled by the conception of nuclear acid therapy and precision medicine. Owing to their amino acid compositions, inherent secondary structures as well as diverse geometrical shapes, peptides or peptide-containing polymers exhibit good biodegradability, high flexibility, and bio-functional diversity as nonviral siRNA vectors. Also, a variety of noncovalent nanocomplexes could be built via self-assembling and electrostatic interactions between cationic peptides and siRNAs. Although the peptide/siRNA nanocomplex-based RNAi therapies, STP705 and MIR-19, are under clinical trials, a guideline addressing the current bottlenecks of peptide/siRNA nanocomplex delivery is in high demand for future research and development. In this review, we present strategies for improving the safety and RNAi efficacy of noncovalent peptide/siRNA nanocomplexes in the treatment of genetic disorders. Through thorough analysis of those RNAi formulations using different delivery strategies, we seek to shed light on the rationale of peptide design and modification in constructing robust siRNA delivery systems, including targeted and co-delivery systems. Based on this, we provide a timely and comprehensive understanding of how to engineer biocompatible and efficient peptide-based siRNA vectors.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Guang Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada; Key Laboratory of Chemical Additives for China National Light Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Nan Liu
- Advanced Materials Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250014, China
| | - Xiaoxia Han
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Feng Zhao
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - P Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada; Advanced Materials Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250014, China.
| |
Collapse
|
11
|
Luong HX, Bui HTP, Tung TT. Application of the All-Hydrocarbon Stapling Technique in the Design of Membrane-Active Peptides. J Med Chem 2022; 65:3026-3045. [PMID: 35112864 DOI: 10.1021/acs.jmedchem.1c01744] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The threats of drug resistance and new emerging pathogens have led to an urgent need to develop alternative treatment therapies. Recently, considerable research efforts have focused on membrane-active peptides (MAPs), a category of peptides in drug discovery with antimicrobial, anticancer, and cell penetration activities that have demonstrated their potential to be multifunctional agents. Nonetheless, natural MAPs have encountered various disadvantages, which mainly include poor bioavailability, the lack of a secondary structure in short peptides, and high production costs for long peptide sequences. Hence, an "all-hydrocarbon stapling system" has been applied to these peptides and proven to effectively stabilize the helical conformations, improving proteolytic resistance and increasing both the potency and the cell permeability. In this review, we summarized and categorized the advances made using this powerful technique in the development of stapled MAPs. Furthermore, outstanding issues and suggestions for future design within each subcategory were thoroughly discussed.
Collapse
Affiliation(s)
- Huy Xuan Luong
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam.,PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Vietnam
| | | | - Truong Thanh Tung
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam.,PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Hanoi 12116, Vietnam
| |
Collapse
|
12
|
Yu Z, Tang H, Cong W, Gao F, Li H, Hu H, Wang X, He S. Hydrocarbon stapling modification of peptide alyteserin-2a: Discovery of novel stapled peptide antitumor agents. J Pept Sci 2022; 28:e3401. [PMID: 34989078 DOI: 10.1002/psc.3401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/24/2021] [Accepted: 12/24/2021] [Indexed: 11/10/2022]
Abstract
Alyteserin-2a (ILGKLLSTAAGLLSNL.NH2 ) is isolated from the skin exudates of midwife toad and has a wide range of biological applications. However, the use of alyteserin-2a as an antitumor agent is limited due to its structural flexibility. In this study, a series of stapled peptides were prepared through hydrocarbon stapling modification without destroying the key residues, and their chemical and biological properties were further evaluated for enhancing the application potential of alyteserin-2a in the field of antitumor drugs development. Among them, alyteserin-2a-Sp3 displayed significant improvement in helicity levels, protease resistance, and antitumor activity compared to that of the template peptide alyteserin-2a, indicating that alyteserin-2a-Sp3 had a potential to become a lead compound for the development of novel antitumor drugs. This study confirms the important effect of hydrocarbon stapling strategy on the secondary structure, hydrolase stability and biological activity of alyteserin-2a.
Collapse
Affiliation(s)
- Ziqiang Yu
- College of Sciences, Shanghai University, China
| | - Hua Tang
- Institute of Translational Medicine, Shanghai University, China
| | - Wei Cong
- Institute of Translational Medicine, Shanghai University, China
| | - Fei Gao
- Institute of Translational Medicine, Shanghai University, China
| | - Huaqiang Li
- Institute of Translational Medicine, Shanghai University, China
| | - Honggang Hu
- Institute of Translational Medicine, Shanghai University, China
| | - Xiaoyan Wang
- Chongqing Health Center for Women and Children, Chongqing, China
| | - Shipeng He
- Institute of Translational Medicine, Shanghai University, China
| |
Collapse
|
13
|
Rinoldi C, Zargarian SS, Nakielski P, Li X, Liguori A, Petronella F, Presutti D, Wang Q, Costantini M, De Sio L, Gualandi C, Ding B, Pierini F. Nanotechnology-Assisted RNA Delivery: From Nucleic Acid Therapeutics to COVID-19 Vaccines. SMALL METHODS 2021; 5:e2100402. [PMID: 34514087 PMCID: PMC8420172 DOI: 10.1002/smtd.202100402] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/04/2021] [Indexed: 05/07/2023]
Abstract
In recent years, the main quest of science has been the pioneering of the groundbreaking biomedical strategies needed for achieving a personalized medicine. Ribonucleic acids (RNAs) are outstanding bioactive macromolecules identified as pivotal actors in regulating a wide range of biochemical pathways. The ability to intimately control the cell fate and tissue activities makes RNA-based drugs the most fascinating family of bioactive agents. However, achieving a widespread application of RNA therapeutics in humans is still a challenging feat, due to both the instability of naked RNA and the presence of biological barriers aimed at hindering the entrance of RNA into cells. Recently, material scientists' enormous efforts have led to the development of various classes of nanostructured carriers customized to overcome these limitations. This work systematically reviews the current advances in developing the next generation of drugs based on nanotechnology-assisted RNA delivery. The features of the most used RNA molecules are presented, together with the development strategies and properties of nanostructured vehicles. Also provided is an in-depth overview of various therapeutic applications of the presented systems, including coronavirus disease vaccines and the newest trends in the field. Lastly, emerging challenges and future perspectives for nanotechnology-mediated RNA therapies are discussed.
Collapse
Affiliation(s)
- Chiara Rinoldi
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Pawel Nakielski
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Xiaoran Li
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Anna Liguori
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
| | - Francesca Petronella
- Institute of Crystallography CNR‐ICNational Research Council of ItalyVia Salaria Km 29.300Monterotondo – Rome00015Italy
| | - Dario Presutti
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Qiusheng Wang
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Marco Costantini
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Luciano De Sio
- Department of Medico‐Surgical Sciences and BiotechnologiesResearch Center for BiophotonicsSapienza University of RomeCorso della Repubblica 79Latina04100Italy
- CNR‐Lab. LicrylInstitute NANOTECArcavacata di Rende87036Italy
| | - Chiara Gualandi
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
- Interdepartmental Center for Industrial Research on Advanced Applications in Mechanical Engineering and Materials TechnologyCIRI‐MAMUniversity of BolognaViale Risorgimento 2Bologna40136Italy
| | - Bin Ding
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Filippo Pierini
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| |
Collapse
|
14
|
Pazo M, Salluce G, Lostalé-Seijo I, Juanes M, Gonzalez F, Garcia-Fandiño R, Montenegro J. Short oligoalanine helical peptides for supramolecular nanopore assembly and protein cytosolic delivery. RSC Chem Biol 2021; 2:503-512. [PMID: 34458796 PMCID: PMC8341679 DOI: 10.1039/d0cb00103a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/07/2020] [Indexed: 01/09/2023] Open
Abstract
In this work we report a rational design strategy for the identification of new peptide prototypes for the non-disruptive supramolecular permeation of membranes and the transport of different macromolecular giant cargos. The approach targets a maximal enhancement of helicity in the presence of membranes with sequences bearing the minimal number of cationic and hydrophobic moieties. The here reported folding enhancement in membranes allowed the selective non-lytic translocation of different macromolecular cargos including giant proteins. The transport of different high molecular weight polymers and functional proteins was demonstrated in vesicles and in cells with excellent efficiency and optimal viability. As a proof of concept, functional monoclonal antibodies were transported for the first time into different cell lines and cornea tissues by exploiting the helical control of a short peptide sequence. This work introduces a rational design strategy that can be employed to minimize the number of charges and hydrophobic residues of short peptide carriers to achieve non-destructive transient membrane permeation and transport of different macromolecules. The helical enhancement of a short oligoalanine peptide scaffold in anionic membranes triggered the supramolecular assembly of a nanopore, which allowed the transport and release of proteins in the cytosol of cells and tissues.![]()
Collapse
Affiliation(s)
- Marta Pazo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Giulia Salluce
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Irene Lostalé-Seijo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Marisa Juanes
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Francisco Gonzalez
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain.,Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS) and Service of Ophthalmology, Complejo Hospitalario Universitario de Santiago 15706 Santiago de Compostela Spain
| | - Rebeca Garcia-Fandiño
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Javier Montenegro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| |
Collapse
|
15
|
Wang H, Dawber RS, Zhang P, Walko M, Wilson AJ, Wang X. Peptide-based inhibitors of protein-protein interactions: biophysical, structural and cellular consequences of introducing a constraint. Chem Sci 2021; 12:5977-5993. [PMID: 33995995 PMCID: PMC8098664 DOI: 10.1039/d1sc00165e] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/07/2021] [Indexed: 12/19/2022] Open
Abstract
Protein-protein interactions (PPIs) are implicated in the majority of cellular processes by enabling and regulating the function of individual proteins. Thus, PPIs represent high-value, but challenging targets for therapeutic intervention. The development of constrained peptides represents an emerging strategy to generate peptide-based PPI inhibitors, typically mediated by α-helices. The approach can confer significant benefits including enhanced affinity, stability and cellular penetration and is ingrained in the premise that pre-organization simultaneously pays the entropic cost of binding, prevents a peptide from adopting a protease compliant β-strand conformation and shields the hydrophilic amides from the hydrophobic membrane. This conceptual blueprint for the empirical design of peptide-based PPI inhibitors is an exciting and potentially lucrative way to effect successful PPI inhibitor drug-discovery. However, a plethora of more subtle effects may arise from the introduction of a constraint that include changes to binding dynamics, the mode of recognition and molecular properties. In this review, we summarise the influence of inserting constraints on biophysical, conformational, structural and cellular behaviour across a range of constraining chemistries and targets, to highlight the tremendous success that has been achieved with constrained peptides alongside emerging design opportunities and challenges.
Collapse
Affiliation(s)
- Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences 5625 Renmin St. Changchun 130022 Jilin China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Nanjing 210023 Jiangsu China
| | - Robert S Dawber
- School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Peiyu Zhang
- School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Martin Walko
- School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Andrew J Wilson
- School of Chemistry, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds Woodhouse Lane Leeds LS2 9JT UK
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences 5625 Renmin St. Changchun 130022 Jilin China
- Department of Applied Chemistry and Engineering, University of Science and Technology of China Hefei 230026 China
| |
Collapse
|
16
|
Kim GC, Cheon DH, Lee Y. Challenge to overcome current limitations of cell-penetrating peptides. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140604. [PMID: 33453413 DOI: 10.1016/j.bbapap.2021.140604] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022]
Abstract
The penetration of biological membranes is a prime obstacle for the delivery of pharmaceutical drugs. Cell-penetrating peptide (CPP) is an efficient vehicle that can deliver various cargos across the biological membranes. Since the discovery, CPPs have been rigorously studied to unveil the underlying penetrating mechanism as well as to exploit CPPs for various biomedical applications. This review will focus on the various strategies to overcome current limitations regarding stability, selectivity, and efficacy of CPPs.
Collapse
Affiliation(s)
- Gyu Chan Kim
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Dae Hee Cheon
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Yan Lee
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
17
|
Ha M, Nam SH, Sim K, Chong SE, Kim J, Kim Y, Lee Y, Nam JM. Highly Efficient Photothermal Therapy with Cell-Penetrating Peptide-Modified Bumpy Au Triangular Nanoprisms using Low Laser Power and Low Probe Dose. NANO LETTERS 2021; 21:731-739. [PMID: 33332127 DOI: 10.1021/acs.nanolett.0c04386] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Photothermal therapy (PTT) exploits nanomaterials with optimal heat conversion and cellular penetration using near-infrared (NIR) laser irradiation. However, current PTT agents suffer from inefficient heat conversion, poor intracellular delivery, and a high dose of probes along with excessive laser irradiation, causing limited therapeutic outcomes. Here, bumpy Au triangular nanoprisms (BATrisms) are developed for increasing the surface area, improving cell penetration, shifting the absorption peak to the NIR region, and enhancing the photothermal conversion efficiency (∼86%). Further, leucine (L)- and lysine (K)-rich cell-penetrating peptides (LK peptides) were employed to largely improve their cellular uptake efficiency. Importantly, a significant in vivo therapeutic efficacy with LK-BATrisms was demonstrated in a triple-negative breast cancer xenograft mice model. A very small dose of LK-BATrism (2.5 μg Au) was enough to exert antitumor efficacy under very low laser power (808 nm, 0.25 W/cm2), causing minimal tissue damages while very efficiently killing cancer cells.
Collapse
Affiliation(s)
- Minji Ha
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - So Hee Nam
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Kyunjong Sim
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Seung-Eun Chong
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Jiyeon Kim
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Yuna Kim
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Yan Lee
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Jwa-Min Nam
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
18
|
Hydrocarbon-Stapled Peptide Based-Nanoparticles for siRNA Delivery. NANOMATERIALS 2020; 10:nano10122334. [PMID: 33255624 PMCID: PMC7760004 DOI: 10.3390/nano10122334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/09/2020] [Accepted: 11/21/2020] [Indexed: 01/22/2023]
Abstract
Small interfering RNAs (siRNAs) are promising molecules for developing new therapies based on gene silencing; however, their delivery into cells remains an issue. In this study, we took advantage of stapled peptide technology that has emerged as a valuable strategy to render natural peptides more structured, resistant to protease degradation and more bioavailable, to develop short carriers for siRNA delivery. From the pool of stapled peptides that we have designed and synthesized, we identified non-toxic vectors that were able to efficiently encapsulate siRNA, transport them into the cell and induce gene silencing. Remarkably, the most efficient stapled peptide (JMV6582), is composed of only eight amino-acids and contains only two cationic charges.
Collapse
|
19
|
Khan MM, Filipczak N, Torchilin VP. Cell penetrating peptides: A versatile vector for co-delivery of drug and genes in cancer. J Control Release 2020; 330:1220-1228. [PMID: 33248708 DOI: 10.1016/j.jconrel.2020.11.028] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/09/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022]
Abstract
Biological barriers hamper the efficient delivery of drugs and genes to targeted sites. Cell penetrating peptides (CPP) have the ability to rapidly internalize across biological membranes. CPP have been effective for delivery of various chemotherapeutic agents used to combat cancer. CPP can enhance delivery of drugs to a targeted site when combined with tumor targeting peptides. CPP can be linked with various cargos like nanoparticles, micelles and liposomes to deliver drugs and genes to the cancer cell. Here, we focus on CPP mediated delivery of drugs to the tumor sites, delivery of genes (siRNA,pDNA) and co-delivery of drugs and genes to combat drug resistance.
Collapse
Affiliation(s)
- Muhammad Muzamil Khan
- Center for Pharmaceutical Biotechnology and Nanomedicines, Northeastern University, Boston, MA 02115, USA; Department of Pharmacy, The Islamia University of Bahawalpur, Pakistan.
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicines, Northeastern University, Boston, MA 02115, USA; Departments of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicines, Northeastern University, Boston, MA 02115, USA; Department of Oncology, Radiotherapy and Plastic Surgery I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
20
|
Nam SH, Jang J, Cheon DH, Chong SE, Ahn JH, Hyun S, Yu J, Lee Y. pH-Activatable cell penetrating peptide dimers for potent delivery of anticancer drug to triple-negative breast cancer. J Control Release 2020; 330:898-906. [PMID: 33152392 DOI: 10.1016/j.jconrel.2020.10.063] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023]
Abstract
We developed a pH-activatable cell-penetrating peptide dimer LH2 with histidine residues, which can penetrate cells, specifically in weak acidic conditions, even at few tens of nanomolar concentrations. LH2 effectively delivered paclitaxel into triple-negative breast cancer cells, MDA-MB-231, via formation of non-covalent complexes (PTX-LH2(M)) or covalent conjugates (PTX-LH2(C)). Moreover, LH2 showed prolonged circulation in the body and enhanced accumulation in tumors. Both PTX-LH2(M) and PTX-LH2(C) showed strong antitumor effects in a triple-negative breast cancer grafted mouse model at an extremely low dosage.
Collapse
Affiliation(s)
- So Hee Nam
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Joomyung Jang
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Dae Hee Cheon
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Seung-Eun Chong
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Joon Hyung Ahn
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Soonsil Hyun
- Department of Chemistry and Education, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jaehoon Yu
- Department of Chemistry and Education, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Yan Lee
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
21
|
Yokoo H, Misawa T, Demizu Y. De Novo Design of Cell-Penetrating Foldamers. CHEM REC 2020; 20:912-921. [PMID: 32463155 DOI: 10.1002/tcr.202000047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022]
Abstract
Cell-penetrating peptides (CPPs) have gained much attention as carriers of hydrophilic molecules, such as drugs, peptides, and nucleic acids, into cells. CPPs are mainly composed of cationic amino acid residues, which play an important role in their intracellular uptake via interactions with acidic groups on cell surfaces. In addition, the secondary structures of CPPs also affect their cell-membrane permeability. Based on this knowledge, a variety of cell-penetrating foldamers (oligomers that form organized secondary structures) have been developed to date. In this account, we describe recent attempts to develop cell-penetrating foldamers containing various building blocks, and their application as DDS carriers.
Collapse
Affiliation(s)
- Hidetomo Yokoo
- Division of Organic Chemistry, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki, Kanagawa, 210-9501, Japan.,Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Yokohama, Kanagawa, 230-0045, Japan
| | - Takashi Misawa
- Division of Organic Chemistry, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki, Kanagawa, 210-9501, Japan
| | - Yosuke Demizu
- Division of Organic Chemistry, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki, Kanagawa, 210-9501, Japan.,Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Yokohama, Kanagawa, 230-0045, Japan
| |
Collapse
|
22
|
Jiao JB, Wang GZ, Hu XL, Zang Y, Maisonneuve S, Sedgwick AC, Sessler JL, Xie J, Li J, He XP, Tian H. Cyclodextrin-Based Peptide Self-Assemblies (Spds) That Enhance Peptide-Based Fluorescence Imaging and Antimicrobial Efficacy. J Am Chem Soc 2020; 142:1925-1932. [PMID: 31884796 DOI: 10.1021/jacs.9b11207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
As a result of their high specificity for their corresponding biological targets, peptides have shown significant potential in a range of diagnostic and therapeutic applications. However, their widespread use has been limited by their minimal cell permeability and stability in biological milieus. We describe here a hepta-dicyanomethylene-4H-pyran appended β-cyclodextrin (DCM7-β-CD) that acts as a delivery enhancing "host" for 1-bromonaphthalene-modified peptides, as demonstrated with peptide probes P1-P4. Interaction between the fluorescent peptides P1-P3 and DCM7-β-CD results in the hierarchical formation of unique supramolecular architectures, which we term supramolecular-peptide-dots (Spds). Each Spd (Spd-1, Spd-2, and Spd-3) was found to facilitate the intracellular delivery of the constituent fluorescent probes (P1-P3), thus allowing spatiotemporal imaging of an apoptosis biomarker (caspase-3) and mitosis. Spd-4, incorporating the antimicrobial peptide P4, was found to provide an enhanced therapeutic benefit against both Gram-positive and Gram-negative bacteria relative to P4 alone. In addition, a fluorescent Spd-4 was prepared, which revealed greater bacterial cellular uptake compared to the peptide alone (P4-FITC) in E. coli. (ATCC 25922) and S. aureus (ATCC 25923). This latter observation supports the suggestion that the Spd platform reported here has the ability to facilitate the delivery of a therapeutic peptide and provides an easy-to-implement strategy for enhancing the antimicrobial efficacy of known therapeutic peptides. The present findings thus serve to highlight a new and effective supramolecular delivery approach that is potentially generalizable to overcome limitations associated with functional peptides.
Collapse
Affiliation(s)
- Jin-Biao Jiao
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China.,Université Paris-Saclay, ENS Paris-Saclay, CNRS, PPSM , 61 av President Wilson , F-94235 Cachan , France
| | - Guan-Zhen Wang
- National Center for Drug Screening, State Key Laboratory of Drug Research Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No. 19A Yuquan Rd. , Beijing 100049 , P. R. China
| | - Xi-Le Hu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , P. R. China
| | - Stéphane Maisonneuve
- Université Paris-Saclay, ENS Paris-Saclay, CNRS, PPSM , 61 av President Wilson , F-94235 Cachan , France
| | - Adam C Sedgwick
- Department of Chemistry , The University of Texas at Austin , 105 East 24th Street-A5300 , Austin , Texas 78712-1224 , United States
| | - Jonathan L Sessler
- Department of Chemistry , The University of Texas at Austin , 105 East 24th Street-A5300 , Austin , Texas 78712-1224 , United States
| | - Juan Xie
- Université Paris-Saclay, ENS Paris-Saclay, CNRS, PPSM , 61 av President Wilson , F-94235 Cachan , France
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Rd. , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No. 19A Yuquan Rd. , Beijing 100049 , P. R. China.,Open Studio for Druggability Research of Marine Natural Products Pilot National Laboratory for Marine Science and Technology (Qingdao) , 1 Wenhai Rd. , Aoshanwei , Jimo, Qingdao 266237 , P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China
| | - He Tian
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Rd. , Shanghai 200237 , P. R. China
| |
Collapse
|
23
|
Kim Y, Hwang S, Khalmuratova R, Kang S, Lee M, Song Y, Park JW, Yu J, Shin HW, Lee Y. α-Helical cell-penetrating peptide-mediated nasal delivery of resveratrol for inhibition of epithelial-to-mesenchymal transition. J Control Release 2019; 317:181-194. [PMID: 31785303 DOI: 10.1016/j.jconrel.2019.11.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 11/25/2022]
Abstract
In the present study, we examined the potential of cell-penetrating peptide (CPP)-based intranasal drug delivery for the treatment of localized nasal diseases. Many charged or non-hydrophobic drugs have difficulty penetrating into the nasal epithelium due to intrinsic membrane impermeability and rapid mucociliary clearance in the nasal cavity. To treat chronic rhinosinusitis with nasal polyps (CRSwNP), one of the most common localized nasal diseases, we conjugated resveratrol (RSV) to an amphiphilic α-helical leucine (L)- and lysine (K)-rich CPP (LK) and intranasally delivered it to the interior of nasal epithelial cells for inhibiting epithelial-to-mesenchymal transition (EMT) caused by hypoxia-inducible factor 1α. The RSV-LK conjugate could penetrate into the nasal epithelium and efficiently inhibit EMT, nasal polyp formation, epithelial disruption, and related inflammation in an eosinophilic CRSwNP mouse model, at 10-fold lower doses and with 3-fold less frequent administration than free RSV. Due to the rapid penetration into the nasal epithelium and the therapeutic effect of the RSV-LK conjugate at much lower doses than free RSV, this CPP-based delivery system, with the ability to overcome the tight nasal epithelial barrier, may provide a new strategy for the treatment of localized nasal diseases without the systemic side effects of cargo drugs.
Collapse
Affiliation(s)
- Yumin Kim
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Soyoung Hwang
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Roza Khalmuratova
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Sunah Kang
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Mingyu Lee
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Youngjun Song
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jong-Wan Park
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea; Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Jaehoon Yu
- Department of Chemistry and Education, College of Education, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun-Woo Shin
- Obstructive Upper airway Research (OUaR) Laboratory, Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea; Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Republic of Korea.
| | - Yan Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
24
|
Kang Z, Ding G, Meng Z, Meng Q. The rational design of cell-penetrating peptides for application in delivery systems. Peptides 2019; 121:170149. [PMID: 31491454 DOI: 10.1016/j.peptides.2019.170149] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022]
Abstract
Cell penetrating peptides (CPPs) play a crucial role in the transportation of bioactive molecules. Although CPPs have been used widely in various delivery systems, further applications of CPPs are hampered by several drawbacks, such as high toxicity, low delivery efficiency, proteolytic instability and poor specificity. To design CPPs with great cell-penetrating ability, physicochemical properties and safety, researchers have tried to develop new methods to overcome the defects of CPPs. Briefly, (1) the side chain of arginine containing the guanidinium group is essential for the facilitation of cellular uptake; (2) the hydrophobic counterion complex around the guanidinium-rich backbone can "coat" the highly cationic structure with lipophilic moieties and act as an activator; (3) the conformation-constrained strategy was pursued to shield the peptide, thereby impeding access of the proteolytic enzyme; (4) targeting strategies can increase cell-type specificity of CPPs. In this review, the above four aspects were discussed in detail.
Collapse
Affiliation(s)
- Ziyao Kang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China
| | - Guihua Ding
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China
| | - Zhao Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China
| | - Qingbin Meng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing, 100850, China; Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, China; Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, 133002, China.
| |
Collapse
|
25
|
Abstract
Approximately 75% of all disease-relevant human proteins, including those involved in intracellular protein-protein interactions (PPIs), are undruggable with the current drug modalities (i.e., small molecules and biologics). Macrocyclic peptides provide a potential solution to these undruggable targets because their larger sizes (relative to conventional small molecules) endow them the capability of binding to flat PPI interfaces with antibody-like affinity and specificity. Powerful combinatorial library technologies have been developed to routinely identify cyclic peptides as potent, specific inhibitors against proteins including PPI targets. However, with the exception of a very small set of sequences, the vast majority of cyclic peptides are impermeable to the cell membrane, preventing their application against intracellular targets. This Review examines common structural features that render most cyclic peptides membrane impermeable, as well as the unique features that allow the minority of sequences to enter the cell interior by passive diffusion, endocytosis/endosomal escape, or other mechanisms. We also present the current state of knowledge about the molecular mechanisms of cell penetration, the various strategies for designing cell-permeable, biologically active cyclic peptides against intracellular targets, and the assay methods available to quantify their cell-permeability.
Collapse
Affiliation(s)
- Patrick G. Dougherty
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, United States
| | - Ashweta Sahni
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, Ohio 43210, United States
| |
Collapse
|
26
|
Reid LM, Verma CS, Essex JW. The role of molecular simulations in understanding the mechanisms of cell-penetrating peptides. Drug Discov Today 2019; 24:1821-1835. [DOI: 10.1016/j.drudis.2019.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/12/2019] [Accepted: 06/17/2019] [Indexed: 01/06/2023]
|
27
|
Xu J, Khan AR, Fu M, Wang R, Ji J, Zhai G. Cell-penetrating peptide: a means of breaking through the physiological barriers of different tissues and organs. J Control Release 2019; 309:106-124. [PMID: 31323244 DOI: 10.1016/j.jconrel.2019.07.020] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022]
Abstract
The selective infiltration of cell membranes and tissue barriers often blocks the entry of most active molecules. This natural defense mechanism prevents the invasion of exogenous substances and limits the therapeutic value of most available molecules. Therefore, it is particularly important to find appropriate ways of membrane translocation and therapeutic agent delivery to its target site. Cell penetrating peptides (CPPs) are a group of short peptides harnessed in this condition, possessing a significant capacity for membrane transduction and could be exploited to transfer various biologically active cargoes into the cells. Since their discovery, CPPs have been employed for delivery of a wide variety of therapeutic molecules to treat various disorders including cranial nerve involvement, ocular inflammation, myocardial ischemia, dermatosis and cancer. The promising results of CPPs-derived therapeutics in various tumor models demonstrated a potential and worthwhile scope of CPPs in chemotherapy. This review describes the detailed description of CPPs and CPPs-assisted molecular delivery against various tissues and organs disorders. An emphasis is focused on summarizing the novel insights and achievements of CPPs in surmounting the natural membrane barriers during the last 5 years.
Collapse
Affiliation(s)
- Jiangkang Xu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Abdur Rauf Khan
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Manfei Fu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Rujuan Wang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China.
| |
Collapse
|
28
|
Polyhistidine facilitates direct membrane translocation of cell-penetrating peptides into cells. Sci Rep 2019; 9:9398. [PMID: 31253836 PMCID: PMC6599048 DOI: 10.1038/s41598-019-45830-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 06/14/2019] [Indexed: 12/27/2022] Open
Abstract
The bovine lactoferricin L6 (RRWQWR) has been previously identified as a novel cell-penetrating peptide (CPP) that is able to efficiently internalize into human cells. L6 interacts with quantum dots (QDs) noncovalently to generate stable L6/QD complexes that enter cells by endocytosis. In this study, we demonstrate a modified L6 (HL6; CHHHHHRRWQWRHHHHHC), in which short polyhistidine peptides are introduced into both flanks of L6, has enhanced cell-penetrating ability in human bronchoalveolar carcinoma A549 cells. The mechanism of cellular uptake of HL6/QD complexes is primarily direct membrane translocation rather than endocytosis. Dimethyl sulfoxide (DMSO), but not pyrenebutyrate (PB), ethanol, oleic acid, or 1,2-benzisothiazol-3(2 H)-one (BIT), slightly enhances HL6-mediated protein transduction efficiency. Neither HL6 nor HL6/QD complexes are cytotoxic to A549 or HeLa cells. These results indicate that HL6 could be a more efficient drug carrier than L6 for biomedical as well as biotechnological applications, and that the function of polyhistidine peptides is critical to CPP-mediated protein transduction.
Collapse
|
29
|
Guarracino DA, Riordan JA, Barreto GM, Oldfield AL, Kouba CM, Agrinsoni D. Macrocyclic Control in Helix Mimetics. Chem Rev 2019; 119:9915-9949. [DOI: 10.1021/acs.chemrev.8b00623] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Danielle A. Guarracino
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Jacob A. Riordan
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Gianna M. Barreto
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Alexis L. Oldfield
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Christopher M. Kouba
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| | - Desiree Agrinsoni
- Department of Chemistry, The College of New Jersey, Ewing, New Jersey 08628, United States
| |
Collapse
|
30
|
Gallo M, Defaus S, Andreu D. 1988-2018: Thirty years of drug smuggling at the nano scale. Challenges and opportunities of cell-penetrating peptides in biomedical research. Arch Biochem Biophys 2018; 661:74-86. [PMID: 30447207 DOI: 10.1016/j.abb.2018.11.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/22/2022]
Abstract
In 1988, two unrelated papers reported the discovery of peptide vectors with innate cell translocation properties, setting the ground for a new area of research that over the years has grown into considerable therapeutic potential. The vectors, named cell-penetrating peptides (CPPs), constitute a now large and diversified family, sharing the extraordinary ability to diffuse unaltered across cell membranes while ferrying diverse associated cargos. Such properties have made CPPs ideal tools for delivery of nucleic acids, proteins and other therapeutic/diagnostic molecules to cells and tissues via covalent conjugation or complexation. This year 2018 marks the 30th anniversary of a peptide research landmark opening new perspectives in drug delivery. Given its vastness, exhaustive coverage of the main features and accomplishments in the CPP field is virtually impossible. Hence this manuscript, after saluting the above 30th jubilee, focuses by necessity on the most recent contributions, providing a comprehensive list of recognized CPPs and their latest-reported applications over the last two years. In addition, it thoroughly reviews three areas of peptide vector research of particular interest to us, namely (i) efficient transport of low-bioavailability drugs into the brain; (ii) CPP-delivered disruptors of G protein-coupled receptor (GPCRs) heteromers related to several disorders, and (iii) CPP-mediated delivery of useful but poorly internalized drugs into parasites.
Collapse
Affiliation(s)
- Maria Gallo
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Sira Defaus
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, 08003 Barcelona, Spain.
| | - David Andreu
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, 08003 Barcelona, Spain.
| |
Collapse
|
31
|
Singh T, Murthy ASN, Yang HJ, Im J. Versatility of cell-penetrating peptides for intracellular delivery of siRNA. Drug Deliv 2018; 25:1996-2006. [PMID: 30799658 PMCID: PMC6319457 DOI: 10.1080/10717544.2018.1543366] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 12/05/2022] Open
Abstract
The plasma membrane is a large barrier to systemic drug delivery into cells, and it limits the efficacy of drug cargo. This issue has been overcome using cell-penetrating peptides (CPPs). CPPs are short peptides (6-30 amino acid residues) that are potentially capable of intracellular penetration to deliver drug molecules. CPPs broadened biomedical applications and provide a means to deliver a range of biologically active molecules, such as small molecules, proteins, imaging agents, and pharmaceutical nanocarriers, across the plasma membrane with high efficacy and low toxicity. This review is focused on the versatility of CPPs and advanced approaches for siRNA delivery.
Collapse
Affiliation(s)
- Tejinder Singh
- Department of Chemical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Akula S. N. Murthy
- Department of Chemical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Hye-Jin Yang
- Department of Chemical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Jungkyun Im
- Department of Chemical Engineering, Soonchunhyang University, Asan, Republic of Korea
| |
Collapse
|
32
|
Bouillon C, Bessin Y, Poncet F, Gary-Bobo M, Dumy P, Barboiu M, Bettache N, Ulrich S. Biomolecular dynamic covalent polymers for DNA complexation and siRNA delivery. J Mater Chem B 2018; 6:7239-7246. [DOI: 10.1039/c8tb01278d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Dynamic covalent polymers made from modified amino acids complex nucleic acids and deliver siRNA in living cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Mihail Barboiu
- IEM
- Adaptive Supramolecular Nanosystems Group
- Université de Montpellier
- CNRS
- ENSCM
| | | | | |
Collapse
|