1
|
Catalina-Hernandez E, Aguilella-Arzo M, Peralvarez-Marin A, Lopez-Martin M. Computational Insights into Membrane Disruption by Cell-Penetrating Peptides. J Chem Inf Model 2025. [PMID: 39823544 DOI: 10.1021/acs.jcim.4c01940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Cell-penetrating peptides (CPPs) can translocate into cells without inducing cytotoxicity. The internalization process implies several steps at different time scales ranging from microseconds to minutes. We combine adaptive Steered Molecular Dynamics (aSMD) with conventional Molecular Dynamics (cMD) to observe nonequilibrium and equilibrium states to study the early mechanisms of peptide-bilayer interaction leading to CPPs internalization. We define three membrane compositions representing bilayer sections, neutral lipids (i.e., upper leaflet), neutral lipids with cholesterol (i.e., hydrophobic core), and neutral/negatively charged lipids with cholesterol (i.e., lower leaflet) to study the energy barriers and disruption mechanisms of Arg9, MAP, and TP2, representing cationic, amphiphilic, and hydrophobic CPPs, respectively. Cholesterol and negatively charged lipids increase the energetic barriers for the peptide-bilayer crossing. TP2 interacts with the bilayer by hydrophobic insertion, while Arg9 disrupts the bilayer by forming transient or stable pores. MAP has shown both behaviors. Collectively, these findings underscore the significance of innovative computational approaches in studying membrane-disruptive peptides and, more specifically, in harnessing their potential for cell penetration.
Collapse
Affiliation(s)
- Eric Catalina-Hernandez
- Unit of Biophysics, Department of Biochemistry and Molecular Biology, Facultat de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
- Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| | - Marcel Aguilella-Arzo
- Laboratory of Molecular Biophysics, Department of Physics, University Jaume I, 12071 Castellon, Spain
| | - Alex Peralvarez-Marin
- Unit of Biophysics, Department of Biochemistry and Molecular Biology, Facultat de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
- Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| | - Mario Lopez-Martin
- Unit of Biophysics, Department of Biochemistry and Molecular Biology, Facultat de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
- Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| |
Collapse
|
2
|
Mathath AV, Chakraborty D. Effect of peptide hydrophilicity on membrane curvature and permeation. J Chem Phys 2024; 161:164105. [PMID: 39440757 DOI: 10.1063/5.0226869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
Using a well-developed reaction coordinate in umbrella sampling, we studied the single peptide permeation through a model cancerous cell membrane, varying the hydrophilicity and the charge of the peptides. Two peptides, melittin and pHD108, were studied. The permeation mechanism differs from a barrel-stave-like mechanism to toroidal pore and vesicle formation based on the number and the placement of the hydrophilic amino acids in the peptide. Membrane curvature changes dynamically as the permeation process occurs. In the case of vesicles, the peptide traverses along a smooth, homogenous pathway, whereas a rugged, steep pathway was found when lipid molecules did not line up along the wall of the membrane (barrel-stave-like mechanism). A mechanism similar to a toroidal pore consists of multiple minima. Higher free energy was found for the permeating terminal containing charged amino acid residues. Vesicle formation was found for pHD108 peptide N-terminal with a maximum membrane thinning effect of 54.4% with free energy cost of 8.20 ± 0.10 kcal mol-1 and pore radius of 2.33 ± 0.07 nm. Insights gained from this study can help to build synthetic peptides for drug delivery.
Collapse
Affiliation(s)
- Anjana V Mathath
- Biophysical and Computational Chemistry Laboratory, Department of Chemistry, National Institute of Technology Karnataka, Surathkal, Mangalore 575 025, Karnataka, India
| | - Debashree Chakraborty
- Biophysical and Computational Chemistry Laboratory, Department of Chemistry, National Institute of Technology Karnataka, Surathkal, Mangalore 575 025, Karnataka, India
| |
Collapse
|
3
|
Lund PM, Kristensen K, Larsen NW, Knuhtsen A, Hansen MB, Hjørringgaard CU, Eriksen AZ, Urquhart AJ, Mortensen KI, Simonsen JB, Andresen TL, Larsen JB. Tuning the double lipidation of salmon calcitonin to introduce a pore-like membrane translocation mechanism. J Colloid Interface Sci 2024; 669:198-210. [PMID: 38713958 DOI: 10.1016/j.jcis.2024.04.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 05/09/2024]
Abstract
A widespread strategy to increase the transport of therapeutic peptides across cellular membranes has been to attach lipid moieties to the peptide backbone (lipidation) to enhance their intrinsic membrane interaction. Efforts in vitro and in vivo investigating the correlation between lipidation characteristics and peptide membrane translocation efficiency have traditionally relied on end-point read-out assays and trial-and-error-based optimization strategies. Consequently, the molecular details of how therapeutic peptide lipidation affects it's membrane permeation and translocation mechanisms remain unresolved. Here we employed salmon calcitonin as a model therapeutic peptide and synthesized nine double lipidated analogs with varying lipid chain lengths. We used single giant unilamellar vesicle (GUV) calcein influx time-lapse fluorescence microscopy to determine how tuning the lipidation length can lead to an All-or-None GUV filling mechanism, indicative of a peptide mediated pore formation. Finally, we used a GUVs-containing-inner-GUVs assay to demonstrate that only peptide analogs capable of inducing pore formation show efficient membrane translocation. Our data provided the first mechanistic details on how therapeutic peptide lipidation affects their membrane perturbation mechanism and demonstrated that fine-tuning lipidation parameters could induce an intrinsic pore-forming capability. These insights and the microscopy based workflow introduced for investigating structure-function relations could be pivotal for optimizing future peptide design strategies.
Collapse
Affiliation(s)
- Philip M Lund
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kasper Kristensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Nanna W Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Astrid Knuhtsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Morten B Hansen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Claudia U Hjørringgaard
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Anne Z Eriksen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Andrew J Urquhart
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kim I Mortensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Jens B Simonsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Thomas L Andresen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Jannik B Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark; DTU Health Tech, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
4
|
Khanna A, Shedge SV, Zuehlsdorff TJ, Isborn CM. Calculating absorption and fluorescence spectra for chromophores in solution with ensemble Franck-Condon methods. J Chem Phys 2024; 161:044121. [PMID: 39077907 DOI: 10.1063/5.0217080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
Accurately modeling absorption and fluorescence spectra for molecules in solution poses a challenge due to the need to incorporate both vibronic and environmental effects, as well as the necessity of accurate excited state electronic structure calculations. Nuclear ensemble approaches capture explicit environmental effects, Franck-Condon methods capture vibronic effects, and recently introduced ensemble-Franck-Condon approaches combine the advantages of both methods. In this study, we present and analyze simulated absorption and fluorescence spectra generated with combined ensemble-Franck-Condon approaches for three chromophore-solvent systems and compare them to standard ensemble and Franck-Condon spectra, as well as to the experiment. Employing configurations obtained from ground and excited state ab initio molecular dynamics, three combined ensemble-Franck-Condon approaches are directly compared to each other to assess the accuracy and relative computational time. We find that the approach employing an average finite-temperature Franck-Condon line shape generates spectra nearly identical to the direct summation of an ensemble of Franck-Condon spectra at one-fourth of the computational cost. We analyze how the spectral simulation method, as well as the level of electronic structure theory, affects spectral line shapes and associated Stokes shifts for 7-nitrobenz-2-oxa-1,3-diazol-4-yl and Nile red in dimethyl sulfoxide and 7-methoxy coumarin-4-acetic acid in methanol. For the first time, our studies show the capability of combined ensemble-Franck-Condon methods for both absorption and fluorescence spectroscopy and provide a powerful tool for simulating linear optical spectra.
Collapse
Affiliation(s)
- Ajay Khanna
- Chemistry and Biochemistry, University of California Merced, Merced, California 95343, USA
| | - Sapana V Shedge
- Chemistry and Biochemistry, University of California Merced, Merced, California 95343, USA
| | - Tim J Zuehlsdorff
- Department of Chemistry, Oregon State University, Corvallis, Oregon 97331, USA
| | - Christine M Isborn
- Chemistry and Biochemistry, University of California Merced, Merced, California 95343, USA
| |
Collapse
|
5
|
Ji K, Yao Y, Gao Y, Huang S, Ma L, Pan Q, Wu J, Zhang W, Chen H, Zhang L. Evaluating the cytotoxicity mechanism of the cell-penetrating peptide TP10 on Jurkat cells. Biochimie 2024; 221:182-192. [PMID: 37922978 DOI: 10.1016/j.biochi.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
TP10, a classic cell-penetrating peptide, shows a high degree of similarity to AMPs in structure. Although TP10 has been widely used in drug delivery, the mechanism underlying its cytotoxicity is yet to be elucidated. Herein, we explored the cell-killing mechanism of TP10 against human leukemia Jurkat cells. TP10 induced necrosis in Jurkat cells via rapid disruption of cell membranes, particularly at high concentrations. Although mitochondria in Jurkat cells were damaged by TP10, mitochondria-mediated apoptosis did not occur, possibly due to intracellular ATP depletion. Necroptosis in TP10-treated Jurkat cells became an alternative route of apoptosis. Our results demonstrate that necrosis and necroptosis rather than apoptosis are involved in the cell-killing mechanism of TP10, which contributes to the understanding of its toxicity.
Collapse
Affiliation(s)
- Kun Ji
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Yufan Yao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yuxuan Gao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Sujie Huang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Ling Ma
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qing Pan
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Jun Wu
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Wei Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, 730000, China.
| | - Hongmei Chen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Lei Zhang
- The First Hospital, The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Shin HJ, Lee BK, Kang HA. Transdermal Properties of Cell-Penetrating Peptides: Applications and Skin Penetration Mechanisms. ACS APPLIED BIO MATERIALS 2024; 7:1-16. [PMID: 38079575 DOI: 10.1021/acsabm.3c00659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Cell-penetrating peptides (CPPs) consist of 5-30 amino acids with intracellular transduction abilities and diverse physicochemical properties, origins, and sequences. Although recent developments in bioinformatics have facilitated the prediction of CPP candidates with the potential for transduction into cells, the mechanisms by which CPPs penetrate cells and various tissues have not yet been elucidated at the molecular interaction level. Recently, the skin-penetrating ability of CPPs has gained wide attention and emerged as a simple and effective strategy for the delivery of macromolecules into the skin. Studies on the skin structure have suggested that the penetration potential of CPPs is based on the molecular interactions and characteristics of the lipid lamellar structure between corneocytes in the stratum corneum. This review provides a brief overview of the general properties, transduction mechanisms, applications, and safety issues of CPPs, focusing on CPPs with transdermal properties, that are currently being used to develop therapeutics and cosmetics.
Collapse
Affiliation(s)
- Hee Je Shin
- ProCell R&D Center, ProCell Therapeutics, Inc., #1009 Ace-Twin Tower II, 273, Digital-ro, Guro-gu, Seoul 08381, Republic of Korea
- Department of Life Science, College of Natural Science, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Byung Kyu Lee
- ProCell R&D Center, ProCell Therapeutics, Inc., #1009 Ace-Twin Tower II, 273, Digital-ro, Guro-gu, Seoul 08381, Republic of Korea
| | - Hyun Ah Kang
- Department of Life Science, College of Natural Science, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
7
|
Bennett AL, Cranford KN, Bates AL, Sabatini CR, Lee HS. A molecular dynamics study of cell-penetrating peptide transportan-10 (TP10): Binding, folding and insertion to transmembrane state in zwitterionic membrane. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184218. [PMID: 37634858 PMCID: PMC10843101 DOI: 10.1016/j.bbamem.2023.184218] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/05/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Transportan 10 (TP10) is a 21-residue, cationic, α-helical cell-penetrating peptide that can be used as a delivery vector for various bioactive molecules. Based on recent confocal microscopy studies, it is believed that TP10 can translocate across neutral lipid membrane passively, possibly as a monomer, without the formation of permanent pore. Here, we performed extensive molecular dynamics (MD) simulations of TP10W (Y3W variant of TP10) to find the microscopic details of binding, folding and insertion of TP10W to transmembrane state in POPC bilayer. Binding study with CHARMM36 force field showed that TP10W initially binds to the membrane surface in unstructured configuration, but it spontaneously folds into α-helical conformation under the lipid head groups. Further insertion of TP10W, changing from a surface bound state to a vertically oriented transmembrane state, was investigated via umbrella simulations. The resulting free energy profile shows a relatively small barrier between two states, suggesting a possible translocation pathway as a monomer. In fact, unbiased simulation of transmembrane TP10W revealed how a charged Lys side chain can move from one leaflet to the other without a significant free energy cost. Finally, we compared the results of TP10W simulations with those of point mutated variants (TP10W-K12A18 and TP10W-K19L) to understand the effect of charge distribution on the peptide. It was observed that such a conservative mutation can cause noticeable changes in the conformations of both surface bound and transmembrane states. The results of present study will be discussed in relation to the experimentally observed activities of TP10W against neutral membrane.
Collapse
Affiliation(s)
- Ashley L Bennett
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, NC 28403, United States of America
| | - Kristen N Cranford
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, NC 28403, United States of America
| | - Austin L Bates
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, NC 28403, United States of America
| | - Christopher R Sabatini
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, NC 28403, United States of America
| | - Hee-Seung Lee
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, NC 28403, United States of America.
| |
Collapse
|
8
|
Lica JJ, Heldt M, Wieczór M, Chodnicki P, Ptaszyńska N, Maciejewska N, Łęgowska A, Brankiewicz W, Gucwa K, Stupak A, Pradhan B, Gitlin-Domagalska A, Dębowski D, Milewski S, Bieniaszewska M, Grabe GJ, Hellmann A, Rolka K. Dual-Activity Fluoroquinolone-Transportan 10 Conjugates Offer Alternative Leukemia Therapy during Hematopoietic Cell Transplantation. Mol Pharmacol 2023; 105:39-53. [PMID: 37977824 DOI: 10.1124/molpharm.123.000735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/01/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023] Open
Abstract
Hematopoietic cell transplantation (HCT) is often considered a last resort leukemia treatment, fraught with limited success due to microbial infections, a leading cause of mortality in leukemia patients. To address this critical issue, we explored a novel approach by synthesizing antileukemic agents containing antibacterial substances. This innovative strategy involves conjugating fluoroquinolone antibiotics, such as ciprofloxacin (CIP) or levofloxacin (LVX), with the cell-penetrating peptide transportan 10 (TP10). Here, we demonstrate that the resultant compounds display promising biologic activities in preclinical studies. These novel conjugates not only exhibit potent antimicrobial effects but are also selective against leukemia cells. The cytotoxic mechanism involves rapid disruption of cell membrane asymmetry leading to membrane damage. Importantly, these conjugates penetrated mammalian cells, accumulating within the nuclear membrane without significant effect on cellular architecture or mitochondrial function. Molecular simulations elucidated the aggregation tendencies of TP10 conjugates within lipid bilayers, resulting in membrane disruption and permeabilization. Moreover, mass spectrometry analysis confirmed efficient reduction of disulfide bonds within TP10 conjugates, facilitating release and activation of the fluoroquinolone derivatives. Intriguingly, these compounds inhibited human topoisomerases, setting them apart from traditional fluoroquinolones. Remarkably, TP10 conjugates generated lower intracellular levels of reactive oxygen species compared with CIP and LVX. The combination of antibacterial and antileukemic properties, coupled with selective cytostatic effects and minimal toxicity toward healthy cells, positions TP10 derivatives as promising candidates for innovative therapeutic approaches in the context of antileukemic HCT. This study highlights their potential in search of more effective leukemia treatments. SIGNIFICANCE STATEMENT: Fluoroquinolones are commonly used antibiotics, while transportan 10 (TP10) is a cell-penetrating peptide (CPP) with anticancer properties. In HCT, microbial infections are the primary cause of illness and death. Combining TP10 with fluoroquinolones enhanced their effects on different cell types. The dual pharmacological action of these conjugates offers a promising proof-of-concept solution for leukemic patients undergoing HCT. Strategically designed therapeutics, incorporating CPPs with antibacterial properties, have the potential to reduce microbial infections in the treatment of malignancies.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Mateusz Heldt
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Milosz Wieczór
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Pawel Chodnicki
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Natalia Ptaszyńska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Natalia Maciejewska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Anna Łęgowska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Wioletta Brankiewicz
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Katarzyna Gucwa
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Anna Stupak
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Bhaskar Pradhan
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Agata Gitlin-Domagalska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Dawid Dębowski
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Sławomir Milewski
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Maria Bieniaszewska
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Grzegorz Jan Grabe
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Andrzej Hellmann
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| | - Krzysztof Rolka
- Department of Regenerative Medicine, Faculty of Medicine, Medical University of Warsaw, Poland (J.J.L.); Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry (M.H., N.M., S.M.) and Department of Physical Chemistry, Faculty of Chemistry, (M.W., P.C.) Gdansk University of Technology, Poland; Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, Poland (J.J.L., N.P., A.Ł., A.G.-D., D.D., K.R.); Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo, Norway (W.B.); Department of Microbiology, Faculty of Biology, University of Gdansk, Poland (K.G.); Polpharma Biologics S.A. Gdansk Science and Technology Park, Poland (A.S.); Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, Poland (B.P.); Medical University of Gdansk, Faculty of Medicine, Department of Hematology and Transplantology, Poland (M.B., A.H.); and Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Poland (G.J.G.)
| |
Collapse
|
9
|
Egberink RO, van Asbeck AH, Boswinkel M, Muradjan G, Dieker J, Brock R. Deciphering Structural Determinants Distinguishing Active from Inactive Cell-Penetrating Peptides for Cytosolic mRNA Delivery. Bioconjug Chem 2023; 34:1822-1834. [PMID: 37733627 PMCID: PMC10587869 DOI: 10.1021/acs.bioconjchem.3c00346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/07/2023] [Indexed: 09/23/2023]
Abstract
The formation of noncovalent complexes by mixing of positively charged polymers with negatively charged oligonucleotides (ONs) is a widely explored concept in nanomedicine to achieve cellular delivery of ONs. Uptake of ON complexes occurs through endocytosis, which then requires release of ON from endosomes. As one type of polymer, cell-penetrating peptides (CPPs) are being used which are peptides of about 8-30 amino acids in length. However, only a few CPPs yield effective cytosolic ON delivery and activity. Several strategies have been devised to increase cellular uptake and enhance endosomal release, among which an increase of osmotic pressure through the so-called proton sponge effect, disruption of membrane integrity through membrane activity, and disulfide-mediated polymerization. Here, we address the relevance of these concepts for mRNA delivery by incorporating structural features into the human lactoferrin-derived CPP, which shows uptake but not delivery. The incorporation of histidines was explored to address osmotic pressure and structural motifs of the delivery-active CPP PepFect14 (PF14) to address membrane disturbance, and finally, the impact of polymerization was explored. Whereas oligomerization increased the stability of polyplexes against heparin-induced decomplexation, neither this approach nor the incorporation of histidine residues to promote a proton-sponge effect yielded activity. Also, the replacement of arginine residues with lysine or ornithine residues, as in PF14, was without effect, even though all polyplexes showed cellular uptake. Ultimately, sufficient activity could only be achieved by transferring amphipathic sequence motifs from PF14 into the hLF context with some benefit of oligomerization demonstrating overarching principles of delivery for CPPs, lipid nanoparticles, and other types of delivery polymers.
Collapse
Affiliation(s)
- Rik Oude Egberink
- Department
of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Alexander H. van Asbeck
- Department
of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Milou Boswinkel
- Department
of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Grigor Muradjan
- Department
of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jürgen Dieker
- Department
of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Roland Brock
- Department
of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department
of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain
| |
Collapse
|
10
|
Shaw P, Klausen M, Lilienkampf A, Bradley M. Fluorophore-Tagged Poly-Lysine RAFT Agents: Controlled Synthesis of Trackable Cell-Penetrating Peptide-Polymers. ACS Macro Lett 2023; 12:1280-1285. [PMID: 37695265 PMCID: PMC10586461 DOI: 10.1021/acsmacrolett.3c00460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
The conjugation of a fluorophore and a variety of cell-penetrating peptides onto a RAFT agent allowed for the synthesis of polymers of defined sizes with quantifiable cell-uptake. Each peptide-RAFT agent was used to polymerize acrylamide, acrylate, and styrene monomers to form high or low molecular weight polymers (here 50 or 7.5 kDa) with the peptide having no influence on the RAFT agent's control. The incorporation of a single fluorophore per polymer chain allowed cellular analysis of the uptake of the size-specific peptide-polymers via flow cytometry and confocal microscopy. The cell-penetrating peptides had a direct effect on the efficiency of polymer uptake for both high and low molecular weight polymers, demonstrating the versatility of the strategy. These "all-in-one", synthetically accessible RAFT agents allow highly controlled preparation of synthetic peptide-polymer conjugates and subsequent quantification of their delivery into cells.
Collapse
Affiliation(s)
- Paige
A. Shaw
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, U.K.
| | - Maxime Klausen
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, U.K.
| | - Annamaria Lilienkampf
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, U.K.
| | - Mark Bradley
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, U.K.
- Precision
Healthcare University Research Institute, Queen Mary University of London, 65-67 New Road, E1 1HH London, U.K.
| |
Collapse
|
11
|
Almeida PF. In Search of a Molecular View of Peptide-Lipid Interactions in Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023. [PMID: 37478368 DOI: 10.1021/acs.langmuir.3c00538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Lipid bilayer membranes are often represented as a continuous nonpolar slab with a certain thickness bounded by two more polar interfaces. Phenomena such as peptide binding to the membrane surface, folding, insertion, translocation, and diffusion are typically interpreted on the basis of this view. In this Perspective, I argue that this membrane representation as a hydrophobic continuum solvent is not adequate to understand peptide-lipid interactions. Lipids are not small compared to membrane-active peptides: their sizes are similar. Therefore, peptide diffusion needs to be understood in terms of free volume, not classical continuum mechanics; peptide solubility or partitioning in membranes cannot be interpreted in terms of hydrophobic mismatch between membrane thickness and peptide length; peptide folding and translocation, often involving cationic peptides, can only be understood if realizing that lipids adapt to the presence of peptides and the membrane may undergo considerable lipid redistribution in the process. In all of those instances, the detailed molecular interactions between the peptide residues and the lipid components are essential to understand the mechanisms involved.
Collapse
Affiliation(s)
- Paulo F Almeida
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| |
Collapse
|
12
|
Sun Z, Huang J, Fishelson Z, Wang C, Zhang S. Cell-Penetrating Peptide-Based Delivery of Macromolecular Drugs: Development, Strategies, and Progress. Biomedicines 2023; 11:1971. [PMID: 37509610 PMCID: PMC10377493 DOI: 10.3390/biomedicines11071971] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Cell-penetrating peptides (CPPs), developed for more than 30 years, are still being extensively studied due to their excellent delivery performance. Compared with other delivery vehicles, CPPs hold promise for delivering different types of drugs. Here, we review the development process of CPPs and summarize the composition and classification of the CPP-based delivery systems, cellular uptake mechanisms, influencing factors, and biological barriers. We also summarize the optimization routes of CPP-based macromolecular drug delivery from stability and targeting perspectives. Strategies for enhanced endosomal escape, which prolong its half-life in blood, improved targeting efficiency and stimuli-responsive design are comprehensively summarized for CPP-based macromolecule delivery. Finally, after concluding the clinical trials of CPP-based drug delivery systems, we extracted the necessary conditions for a successful CPP-based delivery system. This review provides the latest framework for the CPP-based delivery of macromolecular drugs and summarizes the optimized strategies to improve delivery efficiency.
Collapse
Affiliation(s)
- Zhe Sun
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Zvi Fishelson
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Chenhui Wang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
13
|
Espeche JC, Varas R, Maturana P, Cutro AC, Maffía PC, Hollmann A. Membrane permeability and antimicrobial peptides: Much more than just making a hole. Pept Sci (Hoboken) 2023. [DOI: 10.1002/pep2.24305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
14
|
Drexler CI, Cyran JD, Webb LJ. Lipid-Specific Direct Translocation of the Cell-Penetrating Peptide NAF-1 44-67 across Bilayer Membranes. J Phys Chem B 2023; 127:2002-2010. [PMID: 36827970 PMCID: PMC10127249 DOI: 10.1021/acs.jpcb.2c08076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
The cell-penetrating peptide NAF-1 has recently emerged as a promising candidate for selective penetration and destruction of cancer cells. It displays numerous membrane-selective behaviors including cell-specific uptake and organelle-specific degradation. In this work, we explore membrane penetration and translocation of NAF-1 in model lipid bilayer vesicles as a function of lipid identity in zwitterionic phosphatidylcholine lipids mixed with anionic phosphatidylserine, phosphatidylglycerol, and phosphatidic acid lipids. By monitoring the digestion of NAF-1 using the protease trypsin located inside but not outside the vesicles, we determined that the translocation of NAF-1 was significantly enhanced by the presence of phosphatidic acid in the membrane compared to the other three anionic or zwitterionic lipids. These findings were correlated to fluorescence measurements of dansyl-labeled NAF-1, which revealed whether noncovalent interactions between NAF-1 and the bilayer were most stable either at the membrane/solution interface or within the membrane interior. Phosphatidic acid promoted interactions with fatty acid tails, while phosphatidylcholine, phosphatidylserine, and phosphatidylglycerol stabilized interactions with polar lipid headgroups. Interfacial vibrational sum frequency spectroscopy experiments revealed that the phosphate moiety on phosphatidic acid headgroups was better hydrated than on the other three lipids, which helped to shuttle NAF-1 into the hydrophobic region. Our findings demonstrate that permeation does not depend on the net charge on phospholipid lipid headgroups in these model vesicles and suggest a model wherein NAF-1 crosses membranes selectively due to lipid-specific interactions at bilayer surfaces.
Collapse
Affiliation(s)
- Chad I Drexler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jenée D Cyran
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Lauren J Webb
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
15
|
Anselmo S, Sancataldo G, Baiamonte C, Pizzolanti G, Vetri V. Transportan 10 Induces Perturbation and Pores Formation in Giant Plasma Membrane Vesicles Derived from Cancer Liver Cells. Biomolecules 2023; 13:biom13030492. [PMID: 36979427 PMCID: PMC10046094 DOI: 10.3390/biom13030492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/30/2023] Open
Abstract
Continuous progress has been made in the development of new molecules for therapeutic purposes. This is driven by the need to address several challenges such as molecular instability and biocompatibility, difficulties in crossing the plasma membrane, and the development of host resistance. In this context, cell-penetrating peptides (CPPs) constitute a promising tool for the development of new therapies due to their intrinsic ability to deliver therapeutic molecules to cells and tissues. These short peptides have gained increasing attention for applications in drug delivery as well as for their antimicrobial and anticancer activity but the general rules regulating the events involved in cellular uptake and in the following processes are still unclear. Here, we use fluorescence microscopy methods to analyze the interactions between the multifunctional peptide Transportan 10 (TP10) and the giant plasma membrane vesicles (GPMVs) derived from cancer cells. This aims to highlight the molecular mechanisms underlying functional interactions which bring its translocation across the membrane or cytotoxic mechanisms leading to membrane collapse and disruption. The Fluorescence Lifetime Imaging Microscopy (FLIM) method coupled with the phasor approach analysis proved to be the winning choice for following highly dynamic spatially heterogeneous events in real-time and highlighting aspects of such complex phenomena. Thanks to the presented approach, we were able to identify and monitor TP10 translocation into the lumen, internalization, and membrane-induced modifications depending on the peptide concentration regime.
Collapse
Affiliation(s)
- Sara Anselmo
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giuseppe Sancataldo
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Concetta Baiamonte
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giuseppe Pizzolanti
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro", Università degli Studi di Palermo, 90127 Palermo, Italy
| | - Valeria Vetri
- Dipartimento di Fisica e Chimica-Emilio Segré, Università degli Studi di Palermo, 90128 Palermo, Italy
- AteN Center-Advanced Technologies Network Center, Università degli Studi di Palermo, 90128 Palermo, Italy
| |
Collapse
|
16
|
Cruz GS, dos Santos AT, de Brito EHS, Rádis-Baptista G. Cell-Penetrating Antimicrobial Peptides with Anti-Infective Activity against Intracellular Pathogens. Antibiotics (Basel) 2022; 11:1772. [PMID: 36551429 PMCID: PMC9774436 DOI: 10.3390/antibiotics11121772] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are natural or engineered peptide sequences with the intrinsic ability to internalize into a diversity of cell types and simultaneously transport hydrophilic molecules and nanomaterials, of which the cellular uptake is often limited. In addition to this primordial activity of cell penetration without membrane disruption, multivalent antimicrobial activity accompanies some CPPs. Antimicrobial peptides (AMPs) with cell-penetrability exert their effect intracellularly, and they are of great interest. CPPs with antimicrobial activity (CPAPs) comprise a particular class of bioactive peptides that arise as promising agents against difficult-to-treat intracellular infections. This short review aims to present the antibacterial, antiparasitic, and antiviral effects of various cell-penetrating antimicrobial peptides currently documented. Examples include the antimicrobial effects of different CPAPs against bacteria that can propagate intracellularly, like Staphylococcus sp., Streptococcus sp., Chlamydia trachomatis, Escherichia coli, Mycobacterium sp., Listeria sp., Salmonella sp. among others. CPAPs with antiviral effects that interfere with the intracellular replication of HIV, hepatitis B, HPV, and herpes virus. Additionally, CPAPs with activity against protozoa of the genera Leishmania, Trypanosoma, and Plasmodium, the etiological agents of Leishmaniasis, Chagas' Disease, and Malaria, respectively. The information provided in this review emphasizes the potential of multivalent CPAPs, with anti-infective properties for application against various intracellular infections. So far, CPAPs bear a promise of druggability for the translational medical use of CPPs alone or in combination with chemotherapeutics. Moreover, CPAPs could be an exciting alternative for pharmaceutical design and treating intracellular infectious diseases.
Collapse
Grants
- CNPq #305316/2021-4 National Council of Research and Development, the Ministry of Science, Technology, and Inno-vation
- CAPES #88882.454432/2019-01 CNPq #401900/2022-3 Improvement of Higher Education Personnel (CAPES), the Ministry of Education and Culture (MEC), the Federal Government of Brazil, and the National Council of Research and Development, the Ministry of Science, Technology (CNPq), and Innovation (MCTI),
Collapse
Affiliation(s)
- Gabriela Silva Cruz
- Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceara, Fortaleza 60416-030, Brazil
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceara, Fortaleza 60165-081, Brazil
| | - Ariane Teixeira dos Santos
- Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceara, Fortaleza 60416-030, Brazil
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceara, Fortaleza 60165-081, Brazil
| | - Erika Helena Salles de Brito
- Microbiology Laboratory, Institute of Health Sciences, University of International Integration of the Afro-Brazilian Lusophony, Redenção 62790-970, Brazil
| | - Gandhi Rádis-Baptista
- Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceara, Fortaleza 60416-030, Brazil
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceara, Fortaleza 60165-081, Brazil
| |
Collapse
|
17
|
Efficient transdermal delivery of functional protein cargoes by a hydrophobic peptide MTD 1067. Sci Rep 2022; 12:10853. [PMID: 35760980 PMCID: PMC9237094 DOI: 10.1038/s41598-022-14463-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
The skin has a protective barrier against the external environment, making the transdermal delivery of active macromolecules very difficult. Cell-penetrating peptides (CPPs) have been accepted as useful delivery tools owing to their high transduction efficiency and low cytotoxicity. In this study, we evaluated the hydrophobic peptide, macromolecule transduction domain 1067 (MTD 1067) as a CPP for the transdermal delivery of protein cargoes of various sizes, including growth hormone-releasing hexapeptide-6 (GHRP-6), a truncated form of insulin-like growth factor-I (des(1-3)IGF-I), and platelet-derived growth factor BB (PDGF-BB). The MTD 1067-conjugated GHRP-6 (MTD-GHRP-6) was chemically synthesized, whereas the MTD 1067-conjugated des(1-3)IGF-I and PDGF-BB proteins (MTD-des(1-3)IGF-I and MTD-PDGF-BB) were generated as recombinant proteins. All the MTD 1067-conjugated cargoes exhibited biological activities identical or improved when compared to those of the original cargoes. The analysis of confocal microscopy images showed that MTD-GHRP-6, MTD-des(1-3)IGF-I, and MTD-PDGF-BB were detected at 4.4-, 18.8-, and 32.9-times higher levels in the dermis, respectively, compared to the control group without MTD. Furthermore, the MTD 1067-conjugated cargoes did not show cytotoxicity. Altogether, our data demonstrate the potential of MTD 1067 conjugation in developing functional macromolecules for cosmetics and drugs with enhanced transdermal permeability.
Collapse
|
18
|
Antimicrobial Peptides as a Promising Therapeutic Strategy for Neisseria Infections. Curr Microbiol 2022; 79:102. [DOI: 10.1007/s00284-022-02767-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 01/13/2022] [Indexed: 11/03/2022]
|
19
|
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: 2.0] [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
|
20
|
Anselmo S, Sancataldo G, Mørck Nielsen H, Foderà V, Vetri V. Peptide-Membrane Interactions Monitored by Fluorescence Lifetime Imaging: A Study Case of Transportan 10. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:13148-13159. [PMID: 34714654 PMCID: PMC8582253 DOI: 10.1021/acs.langmuir.1c02392] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/19/2021] [Indexed: 06/13/2023]
Abstract
The interest on detailed analysis of peptide-membrane interactions is of great interest in both fundamental and applied sciences as these may relate to both functional and pathogenic events. Such interactions are highly dynamic and spatially heterogeneous, making the investigation of the associated phenomena highly complex. The specific properties of membranes and peptide structural details, together with environmental conditions, may determine different events at the membrane interface, which will drive the fate of the peptide-membrane system. Here, we use an experimental approach based on the combination of spectroscopy and fluorescence microscopy methods to characterize the interactions of the multifunctional amphiphilic peptide transportan 10 with model membranes. Our approach, based on the use of suitable fluorescence reporters, exploits the advantages of phasor plot analysis of fluorescence lifetime imaging microscopy measurements to highlight the molecular details of occurring membrane alterations in terms of rigidity and hydration. Simultaneously, it allows following dynamic events in real time without sample manipulation distinguishing, with high spatial resolution, whether the peptide is adsorbed to or inserted in the membrane.
Collapse
Affiliation(s)
- Sara Anselmo
- Dipartimento
di Fisica e Chimica−Emilio Segré, Università degli Studi di Palermo, Viale delle Scienze ed. 18 90128, Palermo, Italy
| | - Giuseppe Sancataldo
- Dipartimento
di Fisica e Chimica−Emilio Segré, Università degli Studi di Palermo, Viale delle Scienze ed. 18 90128, Palermo, Italy
| | - Hanne Mørck Nielsen
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2 2100, Copenhagen, Denmark
| | - Vito Foderà
- Department
of Pharmacy, University of Copenhagen, Universitetsparken 2 2100, Copenhagen, Denmark
| | - Valeria Vetri
- Dipartimento
di Fisica e Chimica−Emilio Segré, Università degli Studi di Palermo, Viale delle Scienze ed. 18 90128, Palermo, Italy
| |
Collapse
|
21
|
Cell-Penetrating Peptides and Transportan. Pharmaceutics 2021; 13:pharmaceutics13070987. [PMID: 34210007 PMCID: PMC8308968 DOI: 10.3390/pharmaceutics13070987] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/21/2022] Open
Abstract
In the most recent 25–30 years, multiple novel mechanisms and applications of cell-penetrating peptides (CPP) have been demonstrated, leading to novel drug delivery systems. In this review, I present a brief introduction to the CPP area with selected recent achievements. This is followed by a nostalgic journey into the research in my own laboratories, which lead to multiple CPPs, starting from transportan and paving a way to CPP-based therapeutic developments in the delivery of bio-functional materials, such as peptides, proteins, vaccines, oligonucleotides and small molecules, etc.
Collapse
|
22
|
Kabelka I, Vácha R. Advances in Molecular Understanding of α-Helical Membrane-Active Peptides. Acc Chem Res 2021; 54:2196-2204. [PMID: 33844916 DOI: 10.1021/acs.accounts.1c00047] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biological membranes separate the interior of cells or cellular compartments from their outer environments. This barrier function of membranes can be disrupted by membrane-active peptides, some of which can spontaneously penetrate through the membranes or open leaky transmembrane pores. However, the origin of their activity/toxicity is not sufficiently understood for the development of more potent peptides. To this day, there are no design rules that would be generally valid, and the role of individual amino acids tends to be sequence-specific.In this Account, we describe recent progress in understanding the design principles that govern the activity of membrane-active peptides. We focus on α-helical amphiphilic peptides and their ability to (1) translocate across phospholipid bilayers, (2) form transmembrane pores, or (3) act synergistically, i.e., to produce a significantly more potent effect in a mixture than the individual components.We refined the description of peptide translocation using computer simulations and demonstrated the effect of selected residues. Our simulations showed the necessity to explicitly include charged residues in the translocation description to correctly sample the membrane perturbations they can cause. Using this description, we calculated the translocation of helical peptides with and without the kink induced by the proline/glycine residue. The presence of the kink had no effect on the translocation barrier, but it decreased the peptide affinity to the membrane and reduced the peptide stability inside the membrane. Interestingly, the effects were mainly caused by the peptide's increased polarity, not the higher flexibility of the kink.Flexibility plays a crucial role in pore formation and affects distinct pore structures in different ways. The presence of a kink destabilizes barrel-stave pores, because the kink prevents the tight packing of peptides in the bundle, which is characteristic of the barrel-stave structure. In contrast, the kink facilitates the formation of toroidal pores, where the peptides are only loosely arranged and do not need to closely assemble. The exact position of the kink in the sequence further determines the preferred arrangement of peptides in the pore, i.e., an hourglass or U-shaped structure. In addition, we demonstrated that two self-associated (via termini) helical peptides could mimic the behavior of peptides with a helix-kink-helix motif.Finally, we review the recent findings on the peptide synergism of the archetypal mixture of Magainin 2 and PGLa peptides. We focused on a bacterial plasma membrane mimic that contains negatively charged lipids and lipids with negative intrinsic curvature. We showed that the synergistic action of peptides was highly dependent on the lipid composition. When the lipid composition and peptide/lipid ratios were changed, the systems exhibited more complex behavior than just the previously reported pore formation. We observed membrane adhesion, fusion, and even the formation of the sponge phase in this regime. Furthermore, enhanced adhesion/partitioning to the membrane was reported to be caused by lipid-induced peptide aggregation.In conclusion, the provided molecular insight into the complex behavior of membrane-active peptides provides clues for the design and modification of antimicrobial peptides or toxins.
Collapse
Affiliation(s)
- Ivo Kabelka
- CEITEC − Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University Kamenice 5, 625 00 Brno, Czech Republic
| | - Robert Vácha
- CEITEC − Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University Kamenice 5, 625 00 Brno, Czech Republic
- Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37 Brno, Czech Republic
| |
Collapse
|
23
|
Tummillo KM, Hazlett KR. Co-Opting Host Receptors for Targeted Delivery of Bioconjugates-From Drugs to Bugs. Molecules 2021; 26:molecules26051479. [PMID: 33803208 PMCID: PMC7963163 DOI: 10.3390/molecules26051479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 11/16/2022] Open
Abstract
Bioconjugation has allowed scientists to combine multiple functional elements into one biological or biochemical unit. This assembly can result in the production of constructs that are targeted to a specific site or cell type in order to enhance the response to, or activity of, the conjugated moiety. In the case of cancer treatments, selectively targeting chemotherapies to the cells of interest limit harmful side effects and enhance efficacy. Targeting through conjugation is also advantageous in delivering treatments to difficult-to-reach tissues, such as the brain or infections deep in the lung. Bacterial infections can be more selectively treated by conjugating antibiotics to microbe-specific entities; helping to avoid antibiotic resistance across commensal bacterial species. In the case of vaccine development, conjugation is used to enhance efficacy without compromising safety. In this work, we will review the previously mentioned areas in which bioconjugation has created new possibilities and advanced treatments.
Collapse
Affiliation(s)
- Kristen M. Tummillo
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
- Admera Health, South Plainfield, NJ 07080, USA
| | - Karsten R.O. Hazlett
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
- Correspondence: ; Tel.: +1-518-262-2338
| |
Collapse
|
24
|
Pokorny A, Almeida PF. The Antibiotic Peptide Daptomycin Functions by Reorganizing the Membrane. J Membr Biol 2021; 254:97-108. [DOI: 10.1007/s00232-021-00175-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/21/2021] [Indexed: 10/22/2022]
|
25
|
Kabelka I, Brožek R, Vácha R. Selecting Collective Variables and Free-Energy Methods for Peptide Translocation across Membranes. J Chem Inf Model 2021; 61:819-830. [PMID: 33566605 DOI: 10.1021/acs.jcim.0c01312] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The selective permeability of cellular membranes is a crucial property for controlled transport into and out of cells. Molecules that can bypass the cellular machinery and spontaneously translocate across membranes could be used as therapeutics or drug carriers. Peptides are a prominent class of such molecules, which include natural and man-developed antimicrobial and cell-penetrating peptides. However, the necessary peptide properties for translocation remain elusive. Computer simulations could uncover these properties once we have a good collective variable (CV) that accurately describes the translocation process. Here, we developed a new CV, which includes a description of peptide insertion, local membrane deformation, and peptide internal degrees of freedom related to its charged groups. By comparison of CVs, we demonstrated that all these components are necessary for an accurate description of peptide translocation. Moreover, the advantages and disadvantages of three common methods for free-energy calculations with our CV were evaluated using the MARTINI coarse-grained model: umbrella sampling, umbrella sampling with replica exchange, and metadynamics. The developed CV leads to the reliable and effective calculation of the free energy of peptide translocation, and thus, it could be useful in the design of spontaneously translocating peptides.
Collapse
Affiliation(s)
- Ivo Kabelka
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Radim Brožek
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Robert Vácha
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.,Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37 Brno, Czech Republic
| |
Collapse
|
26
|
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
|
27
|
Ptaszyńska N, Gucwa K, Olkiewicz K, Heldt M, Serocki M, Stupak A, Martynow D, Dębowski D, Gitlin-Domagalska A, Lica J, Łęgowska A, Milewski S, Rolka K. Conjugates of Ciprofloxacin and Levofloxacin with Cell-Penetrating Peptide Exhibit Antifungal Activity and Mammalian Cytotoxicity. Int J Mol Sci 2020; 21:ijms21134696. [PMID: 32630159 PMCID: PMC7369900 DOI: 10.3390/ijms21134696] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
Seven conjugates composed of well-known fluoroquinolone antibacterial agents, ciprofloxacin (CIP) or levofloxacin (LVX), and a cell-penetrating peptide transportan 10 (TP10-NH2) were synthesised. The drugs were covalently bound to the peptide via an amide bond, methylenecarbonyl moiety, or a disulfide bridge. Conjugation of fluoroquinolones to TP10-NH2 resulted in congeners demonstrating antifungal in vitro activity against human pathogenic yeasts of the Candida genus (MICs in the 6.25–100 µM range), whereas the components were poorly active. The antibacterial in vitro activity of most of the conjugates was lower than the activity of CIP or LVX, but the antibacterial effect of CIP-S-S-TP10-NH2 was similar to the mother fluoroquinolone. Additionally, for two representative CIP and LVX conjugates, a rapid bactericidal effect was shown. Compared to fluoroquinolones, TP10-NH2 and the majority of its conjugates generated a relatively low level of reactive oxygen species (ROS) in human embryonic kidney cells (HEK293) and human myeloid leukemia cells (HL-60). The conjugates exhibited cytotoxicity against three cell lines, HEK293, HepG2 (human liver cancer cell line), and LLC-PK1 (old male pig kidney cells), with IC50 values in the 10–100 µM range and hemolytic activity. The mammalian toxicity was due to the intrinsic cytoplasmic membrane disruption activity of TP10-NH2 since fluoroquinolones themselves were not cytotoxic. Nevertheless, the selectivity index values of the conjugates, both for the bacteria and human pathogenic yeasts, remained favourable.
Collapse
Affiliation(s)
- Natalia Ptaszyńska
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
| | - Katarzyna Gucwa
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
| | - Katarzyna Olkiewicz
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
| | - Mateusz Heldt
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdańsk, Poland; (M.H.); (M.S.); (D.M.); (S.M.)
| | - Marcin Serocki
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdańsk, Poland; (M.H.); (M.S.); (D.M.); (S.M.)
| | - Anna Stupak
- Laboratory of Bacterial Genetics, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdańsk, Poland;
| | - Dorota Martynow
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdańsk, Poland; (M.H.); (M.S.); (D.M.); (S.M.)
| | - Dawid Dębowski
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
| | - Agata Gitlin-Domagalska
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
| | - Jan Lica
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
- Correspondence:
| | - Anna Łęgowska
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
| | - Sławomir Milewski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdańsk, Poland; (M.H.); (M.S.); (D.M.); (S.M.)
| | - Krzysztof Rolka
- Department of Molecular Biochemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdańsk, Poland; (N.P.); (K.G.); (K.O.); (D.D.); (A.G.-D.); (A.Ł.); (K.R.)
| |
Collapse
|
28
|
Shuma ML, Moghal MMR, Yamazaki M. Detection of the Entry of Nonlabeled Transportan 10 into Single Vesicles. Biochemistry 2020; 59:1780-1790. [DOI: 10.1021/acs.biochem.0c00102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Madhabi Lata Shuma
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan
| | - Md. Mizanur Rahman Moghal
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan
| | - Masahito Yamazaki
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka 422-8529, Japan
- Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka 422-8529, Japan
- Department of Physics, Faculty of Science, Shizuoka University, Shizuoka 422-8529, Japan
| |
Collapse
|
29
|
Liposomal membrane permeability assessment by fluorescence techniques: Main permeabilizing agents, applications and challenges. Int J Pharm 2020; 580:119198. [PMID: 32169353 DOI: 10.1016/j.ijpharm.2020.119198] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/19/2020] [Accepted: 03/02/2020] [Indexed: 01/08/2023]
Abstract
Liposomes are lipid vesicles made of one or multiple lipid bilayers surrounding an internal aqueous core. They are broadly employed as models to study membrane structure and properties. Among these properties, liposome membrane permeability is crucial and widely assessed by fluorescence techniques. The first part of this review is devoted to describe the various techniques used for membrane permeability assessment. Attention is paid to fluorescence techniques based on vesicle leakage of self-quenching probes, dye/quencher pair or cation/ligand pair. Secondly, the membrane-active agents inducing membrane permeabilization is presented and details on their mechanisms of action are given. Emphasis is also laid on the intrinsic and extrinsic factors that can modulate the membrane permeability. Hence, a suitable liposomal membrane should be formulated according to the aim of the study and its application.
Collapse
|
30
|
Chen C, Richter F, Guerrero-Sanchez C, Traeger A, Schubert US, Feng A, Thang SH. Cell-Penetrating, Peptide-Based RAFT Agent for Constructing Penetration Enhancers. ACS Macro Lett 2020; 9:260-265. [PMID: 35638688 DOI: 10.1021/acsmacrolett.9b00647] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peptide-polymer conjugates represent a promising class of compounds that can be used to overcome some of the limitations associated with peptides intended for therapeutic and diagnostic applications. The efficient generation of well-defined peptide/protein-polymer conjugates can promote the development of the design and synthesis of functional drugs and gene delivery platforms. In this research, a sequence defined cell penetrating peptide (i.e., Transportan 10 (TP 10))-based chain transfer agent (TP-CTA) was designed and synthesized in an automated peptide synthesizer. Thereafter, amphiphilic block copolymers poly[oligo(ethylene glycol) methyl ether acrylate]-b-poly(n-butyl acrylate) (TP-POEGA-b-PBA) were synthesized using the TP-CTA via reversible addition-fragmentation chain transfer (RAFT) polymerization. Circular dichroism (CD) spectroscopy confirmed the preservation of α-helix structure of TP 10, which is crucial for its bioactivity. Transmission electron microscopy (TEM) revealed the formation of self-assembled rod-like and vesicle nanostructures in an aqueous environment. Finally, the obtained peptide-conjugated block copolymers were demonstrated to be effective compounds for cell penetration. This method opens up a way for accessing peptide-polymer conjugates with cell-penetrating abilities.
Collapse
Affiliation(s)
- Chao Chen
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering; College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 7743 Jena, Germany
| | - Carlos Guerrero-Sanchez
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 7743 Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 7743 Jena, Germany
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 7743 Jena, Germany
| | - Anchao Feng
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering; College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - San H. Thang
- School of Chemistry, Monash University, Clayton Campus, Victoria 3800, Australia
| |
Collapse
|
31
|
Geng J, Guo X, Wang L, Nguyen RQ, Wang F, Liu C, Wang H. Intracellular Delivery of DNA and Protein by a Novel Cell-Permeable Peptide Derived from DOT1L. Biomolecules 2020; 10:E217. [PMID: 32024261 PMCID: PMC7072583 DOI: 10.3390/biom10020217] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/27/2022] Open
Abstract
Cellular uptake and intracellular release efficiency of biomacromolecules is low because of hurdles in the cell membrane that result in limited access to intra-cellular targets with few functional effects. Cell-penetrating peptides (CPPs) act as cargo delivery vehicles to promote therapeutic molecule translocation. Here, we describe the novel CPP-Dot1l that not only penetrates by itself, but also mediates cargo translocation in cultured cells, as confirmed by fluorescence microscopy and fluorescence spectrophotometry. We conducted cytotoxicity assays and safety evaluations, and determined peptide-membrane interactions to understand the possible pathway for cargo translocation. Additional nucleic acid and covalently conjugated green fluorescence protein (GFP) studies mediated by CPP-Dot1l were conducted to show functional delivery potential. Results indicate that CPP-Dot1l is a novel and effective CPP due to its good penetrating properties in different cell lines and its ability to enter cells in a concentration-dependent manner. Its penetration efficiency can be prompted by DMSO pretreatment. In addition, not only can it mediate plasmid delivery, but CPP-Dot1l can also deliver GFP protein into cytosol. In conclusion, the findings of this study showed CPP-Dot1l is an attractive pharmaceutical and biochemical tool for future drug, regenerative medicine, cell therapy, gene therapy, and gene editing-based therapy development.
Collapse
Affiliation(s)
- Jingping Geng
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Xiangli Guo
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Lidan Wang
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Richard Q. Nguyen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Fengqin Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Changbai Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China;
| | - Hu Wang
- Department of Pathology and Immunology, Medical School, China Three Gorges University, Yichang 443002, China; (J.G.); (X.G.); (L.W.)
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
32
|
Moghal MMR, Islam MZ, Hossain F, Saha SK, Yamazaki M. Role of Membrane Potential on Entry of Cell-Penetrating Peptide Transportan 10 into Single Vesicles. Biophys J 2019; 118:57-69. [PMID: 31810658 DOI: 10.1016/j.bpj.2019.11.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/27/2022] Open
Abstract
Cell-penetrating peptides (CPPs) can translocate across plasma membranes to enter the cytosol of eukaryotic cells without decreasing cell viability. We revealed the mechanism underlying this translocation by examining the effect of membrane potential, φm, on the entry of a CPP, transportan 10 (TP10), into the lumen of single giant unilamellar vesicles (GUVs). For this purpose, we used the single GUV method to detect the entry of carboxyfluorescein (CF)-labeled TP10 (CF-TP10) into the lumen of single GUVs. First, we used various K+ concentration differences to apply different negative membrane potentials on single GUVs containing gramicidin A in their membrane and confirmed these potentials using the φm-sensitive fluorescent probe 3,3'-dihexyloxacarbocyanine iodine. The fluorescence intensity of the GUV membranes (i.e., the rim intensity) due to 3,3'-dihexyloxacarbocyanine iodine increased with |φm| up to 118 mV, and its dependence on |φm| less than 28 mV agreed with a theoretical estimation (i.e., the dye concentration in the inner leaflet of a GUV is larger than that in the outer leaflet according to the Boltzmann distribution). We then examined the effect of φm on the entry of CF-TP10 into GUVs using single GUVs containing small GUVs or large unilamellar vesicles inside the mother GUV lumen. We found that CF-TP10 entered the GUV lumen without pore formation and the rate of entry of CF-TP10 into the GUV lumen, Ventry, increased with an increase in |φm|. The rim intensity due to CF-TP10 increased with an increase in |φm|, indicating that the CF-TP10 concentration in the inner leaflet of the GUV increased with |φm|. These results indicate that the φm-induced elevation in Ventry can be explained by the increase in CF-TP10 concentration in the inner leaflet with |φm|. We discuss the mechanism underlying this effect of membrane potential based on the pre-pore model of the translocation of CF-TP10 across a GUV membrane.
Collapse
Affiliation(s)
- Md Mizanur Rahman Moghal
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan
| | - Md Zahidul Islam
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan
| | - Farzana Hossain
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan
| | - Samiron Kumar Saha
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan
| | - Masahito Yamazaki
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, Japan; Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, Shizuoka, Japan; Department of Physics, Faculty of Science, Shizuoka University, Shizuoka, Japan.
| |
Collapse
|
33
|
Kardani K, Milani A, H Shabani S, Bolhassani A. Cell penetrating peptides: the potent multi-cargo intracellular carriers. Expert Opin Drug Deliv 2019; 16:1227-1258. [PMID: 31583914 DOI: 10.1080/17425247.2019.1676720] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Cell penetrating peptides (CPPs) known as protein translocation domains (PTD), membrane translocating sequences (MTS), or Trojan peptides (TP) are able to cross biological membranes without clear toxicity using different mechanisms, and facilitate the intracellular delivery of a variety of bioactive cargos. CPPs could overcome some limitations of drug delivery and combat resistant strains against a broad range of diseases. Despite delivery of different therapeutic molecules by CPPs, they lack cell specificity and have a short duration of action. These limitations led to design of combined cargo delivery systems and subsequently improvement of their clinical applications. Areas covered: This review covers all our studies and other researchers in different aspects of CPPs such as classification, uptake mechanisms, and biomedical applications. Expert opinion: Due to low cytotoxicity of CPPs as compared to other carriers and final degradation to amino acids, they are suitable for preclinical and clinical studies. Generally, the efficiency of CPPs was suitable to penetrate the cell membrane and deliver different cargos to specific intracellular sites. However, no CPP-based therapeutic approach has approved by FDA, yet; because there are some disadvantages for CPPs including short half-life in blood, and nonspecific CPP-mediated delivery to normal tissue. Thus, some methods were used to develop the functions of CPPs in vitro and in vivo including the augmentation of cell specificity by activatable CPPs, specific transport into cell organelles by insertion of corresponding localization sequences, incorporation of CPPs into multifunctional dendrimeric or liposomal nanocarriers to improve selectivity and efficiency especially in tumor cells.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Samaneh H Shabani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran , Iran
| |
Collapse
|
34
|
Jafari B, Pourseif MM, Barar J, Rafi MA, Omidi Y. Peptide-mediated drug delivery across the blood-brain barrier for targeting brain tumors. Expert Opin Drug Deliv 2019; 16:583-605. [DOI: 10.1080/17425247.2019.1614911] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Behzad Jafari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia,
Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Mohammad A. Rafi
- Department of Neurology, College of Medicine, Thomas Jefferson University, Philadelphia,
PA, USA
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| |
Collapse
|
35
|
Hasan M, Moghal MMR, Saha SK, Yamazaki M. The role of membrane tension in the action of antimicrobial peptides and cell-penetrating peptides in biomembranes. Biophys Rev 2019; 11:431-448. [PMID: 31093936 DOI: 10.1007/s12551-019-00542-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/29/2019] [Indexed: 12/28/2022] Open
Abstract
For antimicrobial peptides (AMPs) with antimicrobial and bactericidal activities and cell-penetrating peptides (CPPs) with activity to permeate through plasma membrane, their interactions with lipid bilayer region in plasma membrane play important roles in these functions. However, the elementary processes and mechanisms of their functions have not been clear. The single giant unilamellar vesicle (GUV) method has revealed the details of elementary processes of interaction of some AMPs and CPPs with lipid vesicles. In this review, we summarize the mode of action of AMPs such as magainin 2 (Mag) and CPPs such as transportan 10 (TP10), revealed by the single GUV methods, and especially we focus on the role of membrane tension in actions of Mag and TP10 and the mechanisms of their actions. First, we explain the characteristics of the single GUV method briefly. Next, we summarize the recent view on the effect of tension on physical properties of lipid bilayers and describe the role of tension in actions of Mag and TP10. Some experimental results indicate that Mag-induced pore is a stretch-activated pore. The effect of packing of transbilayer asymmetric lipid on Mag-induced pore formation is described. On the other hand, entry of fluorescent dye, carboxyfluorescein (CF)-labeled TP10 (i.e., CF-TP10), into single GUVs without pore formation is affected by tension and high concentration of cholesterol. Pre-pore model for translocation of CF-TP10 across lipid bilayer is described. The experimental methods and their analysis described here are useful for investigation of functions of the other types of AMPs, CPPs, and proteins.
Collapse
Affiliation(s)
- Moynul Hasan
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, 422-8529, Japan.,Department of Pharmacy, Jagannath University, Dhaka, 1100, Bangladesh
| | - Md Mizanur Rahman Moghal
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, 422-8529, Japan
| | - Samiron Kumar Saha
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, 422-8529, Japan
| | - Masahito Yamazaki
- Integrated Bioscience Section, Graduate School of Science and Technology, Shizuoka University, Shizuoka, 422-8529, Japan. .,Nanomaterials Research Division, Research Institute of Electronics, Shizuoka University, 836 Oya, Suruga-ku, Shizuoka, 422-8529, Japan. .,Department of Physics, Faculty of Science, Shizuoka University, Shizuoka, 422-8529, Japan.
| |
Collapse
|
36
|
Ruczyński J, Rusiecka I, Turecka K, Kozłowska A, Alenowicz M, Gągało I, Kawiak A, Rekowski P, Waleron K, Kocić I. Transportan 10 improves the pharmacokinetics and pharmacodynamics of vancomycin. Sci Rep 2019; 9:3247. [PMID: 30824786 PMCID: PMC6397271 DOI: 10.1038/s41598-019-40103-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/11/2019] [Indexed: 12/23/2022] Open
Abstract
In the presented study, transportan 10 (TP10), an amphipathic cell penetrating peptide (CPP) with high translocation activity, was conjugated with vancomycin (Van), which is known for poor access to the intracellular bacteria and the brain. The antibacterial activity of the conjugates was tested on selected clinical strains of methicillin-resistant Staphylococcus aureus (MRSA) and Enterococcus sp. It turned out that all of them had superior antimicrobial activity in comparison to that of free Van, which became visible particularly against clinical MRSA strains. Furthermore, one of the conjugates was tested against MRSA - infected human cells. With respect to them, this compound showed high bactericidal activity. Next, the same conjugate was screened for its capacity to cross the blood brain barrier (BBB). Therefore, qualitative and quantitative analyses of the conjugate's presence in the mouse brain slices were carried out after its iv administration. They indicated the conjugate's presence in the brain in amount >200 times bigger than that of Van. The conjugates were safe with respect to erythrocyte toxicity (erythrocyte lysis assay). Van in the form of a conjugate with TP10 acquires superior pharmacodynamic and pharmacokinetic.
Collapse
Affiliation(s)
- Jarosław Ruczyński
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Izabela Rusiecka
- Department of Pharmacology, Medical University of Gdansk, Debowa 23, 80-204, Gdansk, Poland.
| | - Katarzyna Turecka
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Gdansk, Hallera 107, 80-416, Gdansk, Poland
| | - Agnieszka Kozłowska
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Magdalena Alenowicz
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Iwona Gągało
- Department of Pharmacology, Medical University of Gdansk, Debowa 23, 80-204, Gdansk, Poland
| | - Anna Kawiak
- Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland
| | - Piotr Rekowski
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Krzysztof Waleron
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Gdansk, Hallera 107, 80-416, Gdansk, Poland
| | - Ivan Kocić
- Department of Pharmacology, Medical University of Gdansk, Debowa 23, 80-204, Gdansk, Poland
| |
Collapse
|
37
|
Rusiecka I, Ruczyński J, Kozłowska A, Backtrog E, Mucha P, Kocić I, Rekowski P. TP10-Dopamine Conjugate as a Potential Therapeutic Agent in the Treatment of Parkinson's Disease. Bioconjug Chem 2019; 30:760-774. [PMID: 30653302 DOI: 10.1021/acs.bioconjchem.8b00894] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD) is a common progressive neurodegenerative disorder for which the current treatment is not fully satisfactory. One of the major drawbacks of current PD therapy is poor penetration of drugs across the blood-brain barrier (BBB). In recent years, cell-penetrating peptides (CPPs) such as Tat, SynB, or TP10 have gained great interest due to their ability to penetrate cell membranes and to deliver different cargos to their targets including the central nervous system (CNS). However, there is no data with respect to the use of CPPs as drug carriers to the brain for the treatment of PD. In the presented research, the covalent TP10-dopamine conjugate was synthesized and its pharmacological properties were characterized in terms of its ability to penetrate the BBB and anti-parkinsonian activity. The results showed that dopamine (DA) in the form of a conjugate with TP10 evidently gained access to the brain tissue, exhibited low susceptibility to O-methylation reaction by catechol- O-methyltransferase (lower than that of DA), possessed a relatively high affinity to both dopamine D1 and D2 receptors (in the case of D1, a much higher than that of DA), and showed anti-parkinsonian activity (higher than that of l-DOPA) in the MPTP-induced preclinical animal model of PD. The presented results prove that the conjugation of TP10 with DA may be a good starting point for the development of a new strategy for the treatment of PD.
Collapse
Affiliation(s)
- Izabela Rusiecka
- Department of Pharmacology , Medical University of Gdańsk , Dębowa 23 , 80-204 Gdańsk , Poland
| | - Jarosław Ruczyński
- Faculty of Chemistry , University of Gdańsk , Wita Stwosza 63 , 80-308 Gdańsk , Poland
| | - Agnieszka Kozłowska
- Faculty of Chemistry , University of Gdańsk , Wita Stwosza 63 , 80-308 Gdańsk , Poland
| | - Ewelina Backtrog
- Faculty of Chemistry , University of Gdańsk , Wita Stwosza 63 , 80-308 Gdańsk , Poland
| | - Piotr Mucha
- Faculty of Chemistry , University of Gdańsk , Wita Stwosza 63 , 80-308 Gdańsk , Poland
| | - Ivan Kocić
- Department of Pharmacology , Medical University of Gdańsk , Dębowa 23 , 80-204 Gdańsk , Poland
| | - Piotr Rekowski
- Faculty of Chemistry , University of Gdańsk , Wita Stwosza 63 , 80-308 Gdańsk , Poland
| |
Collapse
|
38
|
Tripathi PP, Arami H, Banga I, Gupta J, Gandhi S. Cell penetrating peptides in preclinical and clinical cancer diagnosis and therapy. Oncotarget 2018; 9:37252-37267. [PMID: 30647857 PMCID: PMC6324683 DOI: 10.18632/oncotarget.26442] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023] Open
Abstract
Delivery of imaging reagents and drugs to tumors is essential for cancer diagnosis and therapy. In addition to therapeutic and diagnostic functionalities, peptides have potential benefits such as biocompatibility, ease to synthesize, smaller size, by-passing off-target side effects, and achieving the beneficial effects with lower-administered dosages. A particular type of peptide known as cell penetrating peptides (CPP) have been predominantly studied during last twenty years as they are not only capable to translocate themselves across membranes but also allow carrier drugs to translocate across plasma membrane, by different mechanisms depending on the CPP. This is of great potential importance in drug delivery systems, as the ability to pass across membranes is crucial to many drug delivery systems. In spite of significant progress in design and application of CPP, more investigations are required to further improve their delivery to tumors, with reduced side-effect and enhanced therapeutic efficacy. In this review, we emphasis on current advancements in preclinical and clinical trials based on using CPP for more efficient delivery of anti-cancer drugs and imaging reagents to cancer tissues and individual cells associated with them. We discuss the evolution of the CPPs-based strategies for targeted delivery, their current status and strengths, along with summarizing the role of CPPs in targeted drug delivery. We also discuss some recently reported diagnostic applications of engineered protease-responsive substrates and activable imaging complexes. We highlight the recent clinical trial data by providing a road map for better design of the CPPs for future preclinical and clinical applications.
Collapse
Affiliation(s)
- Prem Prakash Tripathi
- CSIR-Indian Institute of Chemical Biology (CSIR-IICB), Kolkata, India.,IICB-Translational Research Unit of Excellence, Kolkata, India
| | - Hamed Arami
- Molecular Imaging Program at Stanford (MIPS), The James H. Clark Center, Stanford University, Stanford, CA, USA.,Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Ivneet Banga
- Department of Bioengineering, University of Texas, Arlington, TX, USA
| | - Jalaj Gupta
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Sonu Gandhi
- DBT-National Institute of Animal Biotechnology (DBT-NIAB), Hyderabad, India
| |
Collapse
|
39
|
Kabelka I, Vácha R. Optimal Hydrophobicity and Reorientation of Amphiphilic Peptides Translocating through Membrane. Biophys J 2018; 115:1045-1054. [PMID: 30177443 DOI: 10.1016/j.bpj.2018.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 11/28/2022] Open
Abstract
Cell-penetrating and some antimicrobial peptides can translocate across lipid bilayers without disrupting the membrane structure. However, the molecular properties required for efficient translocation are not fully understood. We employed the Metropolis Monte Carlo method together with coarse-grained models to systematically investigate free-energy landscapes associated with the translocation of secondary amphiphilic peptides. We studied α-helical peptides with different length, amphiphilicity, and distribution of hydrophobic content and found a common translocation path consisting of adsorption, tilting, and insertion. In the adsorbed state, the peptides are parallel to the membrane plane, whereas, in the inserted state, the peptides are perpendicular to the membrane. Our simulations demonstrate that, for all tested peptides, there is an optimal ratio of hydrophilic/hydrophobic content at which the peptides cross the membrane the easiest. Moreover, we show that the hydrophobicity of peptide termini has an important effect on the translocation barrier. These results provide general guidance to optimize peptides for use as carriers of molecular cargos or as therapeutics themselves.
Collapse
Affiliation(s)
- Ivo Kabelka
- CEITEC-Central European Institute of Technology, Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Robert Vácha
- CEITEC-Central European Institute of Technology, Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic; Department of Condensed Matter Physics, Faculty of Science, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
40
|
Kim GC, Ahn JH, Oh JH, Nam S, Hyun S, Yu J, Lee Y. Photoswitching of Cell Penetration of Amphipathic Peptides by Control of α-Helical Conformation. Biomacromolecules 2018; 19:2863-2869. [DOI: 10.1021/acs.biomac.8b00428] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Moghal MMR, Islam MZ, Sharmin S, Levadnyy V, Moniruzzaman M, Yamazaki M. Continuous detection of entry of cell-penetrating peptide transportan 10 into single vesicles. Chem Phys Lipids 2018; 212:120-129. [DOI: 10.1016/j.chemphyslip.2018.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/22/2018] [Accepted: 02/05/2018] [Indexed: 01/06/2023]
|
42
|
Hasan M, Karal MAS, Levadnyy V, Yamazaki M. Mechanism of Initial Stage of Pore Formation Induced by Antimicrobial Peptide Magainin 2. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:3349-3362. [PMID: 29446954 DOI: 10.1021/acs.langmuir.7b04219] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Antimicrobial peptide magainin 2 forms pores in lipid bilayers, a property that is considered the main cause of its bactericidal activity. Recent data suggest that tension or stretching of the inner monolayer plays an important role in magainin 2-induced pore formation in lipid bilayers. Here, to elucidate the mechanism of magainin 2-induced pore formation, we investigated the effect on pore formation of asymmetric lipid distribution in two monolayers. First, we developed a method to prepare giant unilamellar vesicles (GUVs) composed of dioleoylphosphatidylglycerol (DOPG), dioleoylphosphatidylcholine (DOPC), and lyso-PC (LPC) in the inner monolayer and of DOPG/DOPC in the outer monolayer. We consider that in these GUVs, the lipid packing in the inner monolayer was larger than that in the outer monolayer. Next, we investigated the interaction of magainin 2 with these GUVs with an asymmetric distribution of LPC using the single GUV method, and found that the rate constant of magainin 2-induced pore formation, kp, decreased with increasing LPC concentration in the inner monolayer. We constructed a quantitative model of magainin 2-induced pore formation, whereby the binding of magainin 2 to the outer monolayer of a GUV induces stretching of the inner monolayer, causing pore formation. A theoretical equation defining kp as a function of magainin 2 surface concentration, X, reasonably explains the experimental relationship between kp and X. This model quantitatively explains the effect on kp of the LPC concentration in the inner monolayer. On the basis of these results, we discuss the mechanism of the initial stage of magainin 2-induced pore formation.
Collapse
Affiliation(s)
- Moynul Hasan
- Integrated Bioscience Section, Graduate School of Science and Technology , Shizuoka University , Shizuoka 422-8529 , Japan
| | - Mohammad Abu Sayem Karal
- Integrated Bioscience Section, Graduate School of Science and Technology , Shizuoka University , Shizuoka 422-8529 , Japan
| | - Victor Levadnyy
- Integrated Bioscience Section, Graduate School of Science and Technology , Shizuoka University , Shizuoka 422-8529 , Japan
- Theoretical Problem Center of Physico-Chemical Pharmacology , Russian Academy of Sciences , Kosugina, 4 , 117977 Moscow , Russia
| | - Masahito Yamazaki
- Integrated Bioscience Section, Graduate School of Science and Technology , Shizuoka University , Shizuoka 422-8529 , Japan
- Nanomaterials Research Division, Research Institute of Electronics , Shizuoka University , 836 Oya , Suruga-ku, Shizuoka 422-8529 , Japan
- Department of Physics, Faculty of Science , Shizuoka University , Shizuoka 422-8529 , Japan
| |
Collapse
|
43
|
Islam MZ, Sharmin S, Moniruzzaman M, Yamazaki M. Elementary processes for the entry of cell-penetrating peptides into lipid bilayer vesicles and bacterial cells. Appl Microbiol Biotechnol 2018. [DOI: 10.1007/s00253-018-8889-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
44
|
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: 4.3] [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
|
45
|
Birch D, Christensen MV, Staerk D, Franzyk H, Nielsen HM. Fluorophore labeling of a cell-penetrating peptide induces differential effects on its cellular distribution and affects cell viability. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2483-2494. [DOI: 10.1016/j.bbamem.2017.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 01/28/2023]
|
46
|
Kreutzberger MA, Pokorny A, Almeida PF. Daptomycin-Phosphatidylglycerol Domains in Lipid Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:13669-13679. [PMID: 29130685 PMCID: PMC5710797 DOI: 10.1021/acs.langmuir.7b01841] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Daptomycin is an acidic, 13-amino acid, cyclic polypeptide that contains a number of nonproteinogenic residues and is modified at its N-terminus with a decanoyl chain. It has been in clinical use since 2003 against selected drug-resistant Staphylococcus aureus and Enterococcus spp infections. In vitro, daptomycin is active against Gram-positive pathogens at low concentrations but its antibiotic activity depends critically on the presence of calcium ions. This dependence has been thought to arise from binding of one or two Ca2+ ions to daptomycin as a required step in its interaction with the bacterial membrane. Here, we investigated the interaction of daptomycin with giant unilamellar vesicles (GUVs) composed 1-palmitoyl-2-oleoylphosphatidylcholine (POPC) and 1-palmitoyl-2-oleoylphosphatidylglycerol (POPG). We used fluorescence confocal microscopy to monitor binding of the peptide to GUVs and follow its effect on the membrane of the vesicle. We found that in the absence of POPG or Ca2+ daptomycin does not bind measurably to the lipid membrane. In the presence of 20-30% PG in the membrane and 2 mM Ca2+, daptomycin induces the formation of membrane domains rich in acidic lipids. This effect is not induced by Ca2+ alone. In addition, daptomycin causes GUV collapse, but it does not translocate across the membrane to the inside of intact POPC/POPG vesicles. We conclude that pore formation is probably not the mechanism by which the peptide functions. On the other hand, we found that daptomycin coclusters with the anionic phospholipid POPG and the fluorescent probes used, leading to extensive formation of daptomycin-POPG domains in the membrane.
Collapse
|
47
|
Charged Antimicrobial Peptides Can Translocate across Membranes without Forming Channel-like Pores. Biophys J 2017; 113:73-81. [PMID: 28700927 DOI: 10.1016/j.bpj.2017.04.056] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/18/2017] [Accepted: 04/26/2017] [Indexed: 01/08/2023] Open
Abstract
How can highly charged, cationic antimicrobial peptides (AMPs) translocate across hydrophobic lipid bilayers despite the prohibitive energetic penalty to do so? A common explanation has been the formation of peptide-lined channels. However, for most AMPs, no structures of membrane pores have been found despite clear evidence of membrane leakage and antimicrobial activity. The study here suggests an alternative and simple reason: for the AMP PGLa from Xenopus laevis (charge +5), such pores are not needed to explain both leakage and peptide translocation. Elevated-temperature multimicrosecond equilibrium simulations at all-atomistic level reveal that peptides spontaneously translocate across the membrane individually on a timescale of tens of microseconds, without forming pores. Both surface-bound peptides and lipids assist in the one-by-one translocation of the charged side chains. Single peptides can remain in a transmembrane orientation for many microseconds, snorkeling some charged residues to one interface and some to the opposite, but without inducing a water channel. Instead of stable pores, short-lived water bridges occur when two or three peptides connect at their termini, allowing both ion translocation and lipid flip-flop via a brushlike mechanism usually involving the C terminus of one peptide. The results here suggest that for some specific antimicrobial and other membrane active peptides, pore formation may not have to be invoked at all to explain peptide translocation and membrane permeabilization, which may explain why no channel structures for them have been determined experimentally.
Collapse
|
48
|
Fuselier T, Wimley WC. Spontaneous Membrane Translocating Peptides: The Role of Leucine-Arginine Consensus Motifs. Biophys J 2017; 113:835-846. [PMID: 28834720 PMCID: PMC5567601 DOI: 10.1016/j.bpj.2017.06.070] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/14/2017] [Accepted: 06/02/2017] [Indexed: 12/30/2022] Open
Abstract
We previously used an orthogonal high-throughput screen to select peptides that spontaneously cross synthetic lipid bilayers without bilayer disruption. Many of the 12-residue spontaneous membrane translocating peptides (SMTPs) selected from the library contained a 5-residue consensus motif, LRLLR in positions 5-9. We hypothesized that the conserved motif could be a necessary and sufficient minimal motif for translocation. To test this and to explore the mechanism of spontaneous membrane translocation, we synthesized seven arginine placement variants of LRLLRWC and compared their membrane partitioning, translocation, and perturbation to one of the parent SMTPs, called "TP2". Several motif variant peptides translocate into synthetic vesicles with rates that are similar to TP2. However, the peptide containing the selected motif, LRLLRWC, was not the fastest; sequence context is also important for translocation efficiency. Although none of these peptides permeabilize bilayers, the motif peptides translocate faster at higher peptide to lipid ratios, suggesting that bilayer perturbation and/or cooperative interactions are important for their translocation. On the other hand, TP2 translocates slower as its concentration is increased, suggesting that TP2 translocates as a monomer and is inhibited by lateral interactions in the membrane. TP2 and the LRLLR motif peptide induce lipid translocation, suggesting that lipids chaperone them across the bilayer. The other motif peptides do not induce lipid flip-flop, suggesting an alternate mechanism. Concatenated motifs translocate slower than the motifs alone. Variants of TP2 with shorter and longer arginine side-chain analogs translocate slower than TP2. In summary, these results suggest that multiple patterns of leucine and arginine can support spontaneous membrane translocation, and that sequence context is important for the contribution of the motifs. Because motifs do not make simple, additive contributions to spontaneous translocation, rational engineering of novel SMTPs will remain difficult, providing even more reason to pursue SMTP discovery with synthetic molecular evolution.
Collapse
Affiliation(s)
- Taylor Fuselier
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana
| | - William C Wimley
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
49
|
Ablan FDO, Spaller BL, Abdo KI, Almeida PF. Charge Distribution Fine-Tunes the Translocation of α-Helical Amphipathic Peptides across Membranes. Biophys J 2017; 111:1738-1749. [PMID: 27760360 DOI: 10.1016/j.bpj.2016.08.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/09/2016] [Accepted: 08/30/2016] [Indexed: 11/29/2022] Open
Abstract
Hundreds of cationic antimicrobial and cell-penetrating peptides (CPPs) form amphipathic α-helices when bound to lipid membranes. Here, we test two hypotheses for the differences in the ability of these peptides to translocate across membranes. The first, which we now call the hydrophobicity hypothesis, is that peptide translocation is determined by the Gibbs energy of insertion into the bilayer from the membrane interface. The second, which we call the charge-distribution hypothesis, is that translocation is determined by whether the distribution of cationic residues in the peptide can transiently stabilize a high-energy inserted intermediate by forming salt bridges to the phosphates of lipid headgroups. To test these hypotheses, we measured translocation of two series of peptide variants. The first series was based on TP10W, a peptide derived from the amphipathic CPP transportan 10; the second was based on DL1a, a synthetic peptide derived from staphylococcal δ-lysin. The peptides in those two series had small sequence changes relative to TP10W and DL1a: either single-residue substitutions or two-residue switches, which were designed to increase or decrease translocation differently according to the two hypotheses. We found that with regard to the changes introduced in the sequences, five out of six peptide variants translocated in agreement with the charge-distribution hypothesis, whereas none showed agreement with the hydrophobicity hypothesis. We conclude that large effects on translocation are probably determined by hydrophobicity, but the fine tuning appears to arise from the distribution of cationic residues along the peptide sequence.
Collapse
Affiliation(s)
- Francis D O Ablan
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina
| | - B Logan Spaller
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina
| | - Kaitlyn I Abdo
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina
| | - Paulo F Almeida
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina.
| |
Collapse
|
50
|
Patra S, Roy E, Madhuri R, Sharma PK. The next generation cell-penetrating peptide and carbon dot conjugated nano-liposome for transdermal delivery of curcumin. Biomater Sci 2017; 4:418-29. [PMID: 26631310 DOI: 10.1039/c5bm00433k] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To overcome the problems associated with conventional liposomes in transdermal drug delivery like limited penetration ability and poor stability, in this article we report a new generation of cell penetrating peptide polyarginine containing nano-liposomes conjugated with carbon dots. The newly synthesized, cost-effective liposomic precursors were used for the fabrication of liposomes. The resulting liposomes have a bilayer structure like that of conventional liposomes with much smaller size, higher stability, and high penetration ability. The nano-liposomes show high stability at room temperature for three months without any change in size or encapsulation efficiency. The incorporation of carbon dots also opens up their application in fluorescence cell imaging studies, which is very well supported by the fluorescence microscopic analysis of the liposome skin penetration. The as-prepared nano-liposomes do not show any cytotoxicity for MCF-7 cells, even at high concentrations; however, when drug loaded liposomes are applied, they can kill the cancer cells with a high rate. The synthesized nano-liposomes have the potential to be used as an efficient, stable, biocompatible nanocarrier for transdermal drug delivery.
Collapse
Affiliation(s)
- Santanu Patra
- Department of Applied Chemistry, Indian School of Mines, Dhanbad, Jharkhand 826 004, India.
| | - Ekta Roy
- Department of Applied Chemistry, Indian School of Mines, Dhanbad, Jharkhand 826 004, India.
| | - Rashmi Madhuri
- Department of Applied Chemistry, Indian School of Mines, Dhanbad, Jharkhand 826 004, India.
| | - Prashant K Sharma
- Functional Nanomaterials Research Laboratory, Department of Applied Physics, Indian School of Mines, Dhanbad, Jharkhand 826 004, India
| |
Collapse
|