1
|
Morgan DC, Knutson SD, Pan CR, MacMillan DWC. Temporal Microenvironment Mapping (μMap) of Intracellular Trafficking Pathways of Cell-Penetrating Peptides Across the Blood-Brain Barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633151. [PMID: 39868165 PMCID: PMC11761369 DOI: 10.1101/2025.01.15.633151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Peptides play critical roles in cellular functions such as signaling and immune regulation, and peptide-based biotherapeutics show great promise for treating various diseases. Among these, cell-penetrating peptides (CPPs) are particularly valuable for drug delivery due to their ability to cross cell membranes. However, the mechanisms underlying CPP-mediated transport, especially across the blood-brain barrier (BBB), remain poorly understood. Mapping intracellular CPP pathways is essential for advancing drug delivery systems, particularly for neurological disorders, as understanding how CPPs navigate the complex environment of the BBB could enable the development of more effective brain-targeted therapies. Here, we leverage a nanoscale proximity labeling technique, termed μMap, to precisely probe the peptide-receptor interactions and intracellular trafficking mechanisms of photocatalyst-tagged CPPs. The unique advantage of the μMap platform lies in the ability to control the timing of light exposure, which enables the collection of time-gated data, depending on when the blue light is applied to the cells. By harnessing this spatiotemporal precision, we can uncover key peptide-receptor interactions and cellular processes, setting the stage for new innovations in drug design and brain-targeted therapies.
Collapse
Affiliation(s)
- Danielle C Morgan
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Steve D Knutson
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Chenmengxiao Roderick Pan
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - David W C MacMillan
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| |
Collapse
|
2
|
Nguyen SV, Planalp RP, Vashisth H. Role of sequence length and functionalization in interactions of bioconjugated peptides with mitomembranes. Biointerphases 2025; 20:011006. [PMID: 39998173 PMCID: PMC11906191 DOI: 10.1116/6.0004197] [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: 11/10/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Cell-penetrating peptides are efficient tools for intracellular delivery of a variety of cargoes. In this study, we explored the effect of chain length, side chain chemistry, and the locations of conjugated molecules on the interaction between iron-chelating peptides and a mitochondrial-mimicking membrane. We report that a longer chain length enhanced peptide/membrane interactions, and conjugation at the N-terminus lowered the free-energy barrier for peptide translocation across the membrane. Peptides containing Phe side chains and those containing modified Phe (cyclohexane) side chains showed comparable peptide/membrane energetics and translocation energy barriers. Using steered molecular dynamics (SMD) simulations, we further probed the mechanistic details of translocation of each N-terminated peptide across the membrane and compared their metastable states. At a higher steering velocity, the peptide adopted a compact structure due to frequent π-π interactions among conjugated molecules, but at lower steering velocities, each N-terminated peptide adopted an extended structure. This structure allowed cationic residues to maximize their interactions with phosphate headgroups in the mitomembrane. The hydrophobic residues also formed interactions with the lipid acyl tails, facilitating the passage of peptides across the membrane with decreased free energy barriers. Our results highlight the significance of peptide chain length and conjugation in facilitating peptide transport across the membrane.
Collapse
Affiliation(s)
- Son V Nguyen
- Department of Chemistry, University of New Hampshire, Durham, New Hampshire 03824
| | - Roy P Planalp
- Department of Chemistry, University of New Hampshire, Durham, New Hampshire 03824
| | - Harish Vashisth
- Department of Chemistry, University of New Hampshire, Durham, New Hampshire 03824
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire 03824
- Integrated Applied Mathematics Program, University of New Hampshire, Durham, New Hampshire 03824
- Molecular and Cellular Biotechnology Program, University of New Hampshire, Durham, New Hampshire 03824
| |
Collapse
|
3
|
Lopuszynski J, Wang J, Zahid M. Beyond Transduction: Anti-Inflammatory Effects of Cell Penetrating Peptides. Molecules 2024; 29:4088. [PMID: 39274936 PMCID: PMC11397606 DOI: 10.3390/molecules29174088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
One of the bottlenecks to bringing new therapies to the clinic has been a lack of vectors for delivering novel therapeutics in a targeted manner. Cell penetrating peptides (CPPs) have received a lot of attention and have been the subject of numerous developments since their identification nearly three decades ago. Known for their transduction abilities, they have generally been considered inert vectors. In this review, we present a schema for their classification, highlight what is known about their mechanism of transduction, and outline the existing literature as well as our own experience, vis a vis the intrinsic anti-inflammatory properties that certain CPPs exhibit. Given the inflammatory responses associated with viral vectors, CPPs represent a viable alternative to such vectors; furthermore, the anti-inflammatory properties of CPPs, mostly through inhibition of the NF-κB pathway, are encouraging. Much more work in relevant animal models, toxicity studies in large animal models, and ultimately human trials are needed before their potential is fully realized.
Collapse
Affiliation(s)
| | | | - Maliha Zahid
- Department of Cardiovascular Medicine, Guggenheim Gu 9-01B, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
4
|
Gori A, Lodigiani G, Colombarolli SG, Bergamaschi G, Vitali A. Cell Penetrating Peptides: Classification, Mechanisms, Methods of Study, and Applications. ChemMedChem 2023; 18:e202300236. [PMID: 37389978 DOI: 10.1002/cmdc.202300236] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
Cell-penetrating peptides (CPPs) encompass a class of peptides that possess the remarkable ability to cross cell membranes and deliver various types of cargoes, including drugs, nucleic acids, and proteins, into cells. For this reason, CPPs are largely investigated in drug delivery applications in the context of many diseases, such as cancer, diabetes, and genetic disorders. While sharing this functionality and some common structural features, such as a high content of positively charged amino acids, CPPs represent an extremely diverse group of elements, which can differentiate under many aspects. In this review, we summarize the most common characteristics of CPPs, introduce their main distinctive features, mechanistic aspects that drive their function, and outline the most widely used techniques for their structural and functional studies. We highlight current gaps and future perspectives in this field, which have the potential to significantly impact the future field of drug delivery and therapeutics.
Collapse
Affiliation(s)
- Alessandro Gori
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Giulia Lodigiani
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Stella G Colombarolli
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, L.go F. Vito 1, 00168, Roma, Italy
| | - Greta Bergamaschi
- SCITEC - Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, Via Mario Bianco 9, 20131, Milano, Italy
| | - Alberto Vitali
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", National Research Council of Italy, L.go F. Vito 1, 00168, Roma, Italy
| |
Collapse
|
5
|
Gareev K, Tagaeva R, Bobkov D, Yudintceva N, Goncharova D, Combs SE, Ten A, Samochernych K, Shevtsov M. Passing of Nanocarriers across the Histohematic Barriers: Current Approaches for Tumor Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1140. [PMID: 37049234 PMCID: PMC10096980 DOI: 10.3390/nano13071140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
Over the past several decades, nanocarriers have demonstrated diagnostic and therapeutic (i.e., theranostic) potencies in translational oncology, and some agents have been further translated into clinical trials. However, the practical application of nanoparticle-based medicine in living organisms is limited by physiological barriers (blood-tissue barriers), which significantly hampers the transport of nanoparticles from the blood into the tumor tissue. This review focuses on several approaches that facilitate the translocation of nanoparticles across blood-tissue barriers (BTBs) to efficiently accumulate in the tumor. To overcome the challenge of BTBs, several methods have been proposed, including the functionalization of particle surfaces with cell-penetrating peptides (e.g., TAT, SynB1, penetratin, R8, RGD, angiopep-2), which increases the passing of particles across tissue barriers. Another promising strategy could be based either on the application of various chemical agents (e.g., efflux pump inhibitors, disruptors of tight junctions, etc.) or physical methods (e.g., magnetic field, electroporation, photoacoustic cavitation, etc.), which have been shown to further increase the permeability of barriers.
Collapse
Affiliation(s)
- Kamil Gareev
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia
| | - Ruslana Tagaeva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Danila Bobkov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Daria Goncharova
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
| | - Artem Ten
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Konstantin Samochernych
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| |
Collapse
|
6
|
Trofimenko E, Homma Y, Fukuda M, Widmann C. The endocytic pathway taken by cationic substances requires Rab14 but not Rab5 and Rab7. Cell Rep 2021; 37:109945. [PMID: 34731620 DOI: 10.1016/j.celrep.2021.109945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/23/2021] [Accepted: 10/13/2021] [Indexed: 02/01/2023] Open
Abstract
Endocytosis and endosome dynamics are controlled by proteins of the small GTPase Rab family. Besides possible recycling routes to the plasma membrane and various organelles, previously described endocytic pathways (e.g., clathrin-mediated endocytosis, macropinocytosis, CLIC/GEEC pathway) all appear to funnel the endocytosed material to Rab5-positive early endosomes that then mature into Rab7-positive late endosomes/lysosomes. By studying the uptake of a series of cell-penetrating peptides (CPPs), we identify an endocytic pathway that moves material to nonacidic Lamp1-positive late endosomes. Trafficking via this endocytic route is fully independent of Rab5 and Rab7 but requires the Rab14 protein. The pathway taken by CPPs differs from the conventional Rab5-dependent endocytosis at the stage of vesicle formation already, as it is not affected by a series of compounds that inhibit macropinocytosis or clathrin-mediated endocytosis. The Rab14-dependent pathway is also used by physiological cationic molecules such as polyamines and homeodomains found in homeoproteins.
Collapse
Affiliation(s)
- Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
7
|
Obitz D, Miller RG, Metzler-Nolte N. Synthesis and DNA interaction studies of Ru(II) cell penetrating peptide (CPP) bioconjugates. Dalton Trans 2021; 50:13768-13777. [PMID: 34549219 DOI: 10.1039/d1dt01776d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The synthesis of the first bioconjugates of a set of ruthenium(II) dipyridophenazine complexes with two different cell penetrating peptides (CPPs) is described. The CPPs, an arginine rich TAT-9 (RKKRRQRRR) sequence and the Xentry peptide (LCLRPVG), were synthesized using standard SPPS protocols, and the bioconjugates were obtained by the microwave-assisted coupling of the HOBt/TBTU preactivated metal complexes with the respective peptides on Wang resin. The racemic metal complexes were obtained by modified literature procedures. The bioconjugates were cleaved from the resin, purified by semi-preparative HPLC and characterized by analytical HPLC, high resolution mass spectrometry (HR-MS), and NMR spectroscopy. Despite the bioconjugation of the peptides to the dppz ligand, DNA intercalation was observed by CD spectroscopy, viscometry and the characteristic switch-on fluorescence of this class of compounds. Furthermore, the cellular uptake of the Xentry bioconjugates was confirmed by live cell imaging. Like the parent metal complexes, the bioconjugates show low in vitro cytotoxicity (IC50 > 80 μM), which is similar to the respective metal complexes alone.
Collapse
Affiliation(s)
- Daniel Obitz
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany.
| | - Reece G Miller
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany.
| | - Nils Metzler-Nolte
- Inorganic Chemistry I - Bioinorganic Chemistry, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany.
| |
Collapse
|
8
|
Stiltner J, McCandless K, Zahid M. Cell-Penetrating Peptides: Applications in Tumor Diagnosis and Therapeutics. Pharmaceutics 2021; 13:pharmaceutics13060890. [PMID: 34204007 PMCID: PMC8232808 DOI: 10.3390/pharmaceutics13060890] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 01/27/2023] Open
Abstract
Since their identification over twenty-five years ago, the plethora of cell-penetrating peptides (CPP) and their applications has skyrocketed. These 5 to 30 amino acid in length peptides have the unique property of breaching the cell membrane barrier while carrying cargoes larger than themselves into cells in an intact, functional form. CPPs can be conjugated to fluorophores, activatable probes, radioisotopes or contrast agents for imaging tissues, such as tumors. There is no singular mechanism for translocation of CPPs into a cell, and therefore, many CPPs are taken up by a multitude of cell types, creating the challenge of tumor-specific translocation and hindering clinical effectiveness. Varying strategies have been developed to combat this issue and enhance their diagnostic potential by derivatizing CPPs for better targeting by constructing specific cell-activated forms. These methods are currently being used to image integrin-expressing tumors, breast cancer cells, human histiocytic lymphoma and protease-secreting fibrosarcoma cells, to name a few. Additionally, identifying safe, effective therapeutics for malignant tumors has long been an active area of research. CPPs can circumvent many of the complications found in treating cancer with conventional therapeutics by targeted delivery of drugs into tumors, thereby decreasing off-target side effects, a feat not achievable by currently employed conventional chemotherapeutics. Myriad types of chemotherapeutics such as tyrosine kinase inhibitors, antitumor antibodies and nanoparticles can be functionally attached to these peptides, leading to the possibility of delivering established and novel cancer therapeutics directly to tumor tissue. While much research is needed to overcome potential issues with these peptides, they offer a significant advancement over current mechanisms to treat cancer. In this review, we present a brief overview of the research, leading to identification of CPPs with a comprehensive state-of-the-art review on the role of these novel peptides in both cancer diagnostics as well as therapeutics.
Collapse
Affiliation(s)
| | | | - Maliha Zahid
- Correspondence: ; Tel.: +1-412-692-8893; Fax: 412-692-6184
| |
Collapse
|
9
|
Gehan P, Kulifaj S, Soule P, Bodin J, Amoura M, Walrant A, Sagan S, Thiam A, Ngo K, Vivier V, Cribier S, Rodriguez N. Penetratin translocation mechanism through asymmetric droplet interface bilayers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183415. [DOI: 10.1016/j.bbamem.2020.183415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/06/2020] [Accepted: 07/11/2020] [Indexed: 01/15/2023]
|
10
|
Liu J, Fraire JC, De Smedt SC, Xiong R, Braeckmans K. Intracellular Labeling with Extrinsic Probes: Delivery Strategies and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000146. [PMID: 32351015 DOI: 10.1002/smll.202000146] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 06/11/2023]
Abstract
Extrinsic probes have outstanding properties for intracellular labeling to visualize dynamic processes in and of living cells, both in vitro and in vivo. Since extrinsic probes are in many cases cell-impermeable, different biochemical, and physical approaches have been used to break the cell membrane barrier for direct delivery into the cytoplasm. In this Review, these intracellular delivery strategies are discussed, briefly explaining the mechanisms and how they are used for live-cell labeling applications. Methods that are discussed include three biochemical agents that are used for this purpose-purpose-different nanocarriers, cell penetrating peptides and the pore-foraming bacterial toxin streptolysin O. Most successful intracellular label delivery methods are, however, based on physical principles to permeabilize the membrane and include electroporation, laser-induced photoporation, micro- and nanoinjection, nanoneedles or nanostraws, microfluidics, and nanomachines. The strengths and weaknesses of each strategy are discussed with a systematic comparison provided. Finally, the extrinsic probes that are reported for intracellular labeling so-far are summarized, together with the delivery strategies that are used and their performance. This combined information should provide for a useful guide for choosing the most suitable delivery method for the desired probes.
Collapse
Affiliation(s)
- Jing Liu
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Juan C Fraire
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent, B-9000, Belgium
- Joint Laboratory of Advanced Biomedical Technology (NFU-UGent), College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, 210037, P. R. China
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent, B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent, B-9000, Belgium
| |
Collapse
|
11
|
Day AH, Übler MH, Best HL, Lloyd-Evans E, Mart RJ, Fallis IA, Allemann RK, Al-Wattar EAH, Keymer NI, Buurma NJ, Pope SJA. Targeted cell imaging properties of a deep red luminescent iridium(iii) complex conjugated with a c-Myc signal peptide. Chem Sci 2020; 11:1599-1606. [PMID: 32206278 PMCID: PMC7069228 DOI: 10.1039/c9sc05568a] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/14/2019] [Indexed: 12/05/2022] Open
Abstract
A nuclear localisation sequence (NLS) peptide, PAAKRVKLD, derived from the human c-Myc regulator gene, has been functionalised with a long wavelength (λ ex = 550 nm; λ em = 677 nm) cyclometalated organometallic iridium(iii) complex to give the conjugate Ir-CMYC. Confocal fluorescence microscopy studies on human fibroblast cells imaged after 18-24 h incubation show that Ir-CMYC concentrations of 80-100 μM promote good cell uptake and nuclear localisation, which was confirmed though co-localisation studies using Hoechst 33342. In comparison, a structurally related, photophysically analogous iridium(iii) complex lacking the peptide sequence, Ir-PYR, showed very different biological behaviour, with no evidence of nuclear, lysosomal or autophagic vesicle localisation and significantly increased toxicity to the cells at concentrations >10 μM that induced mitochondrial dysfunction. Supporting UV-visible and circular dichroism spectroscopic studies show that Ir-PYR and Ir-CMYC display similarly low affinities for DNA (ca. 103 M-1), consistent with electrostatic binding. Therefore the translocation and nuclear uptake properties of Ir-CMYC are attributed to the presence of the PAAKRVKLD nuclear localisation sequence in this complex.
Collapse
Affiliation(s)
- Adam H Day
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Martin H Übler
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Hannah L Best
- School of Biosciences , Cardiff University , Sir Martin Evans Building , Cardiff , UK
| | - Emyr Lloyd-Evans
- School of Biosciences , Cardiff University , Sir Martin Evans Building , Cardiff , UK
| | - Robert J Mart
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Ian A Fallis
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Rudolf K Allemann
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Eman A H Al-Wattar
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Nathaniel I Keymer
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Niklaas J Buurma
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| | - Simon J A Pope
- School of Chemistry , Cardiff University , Main Building , Cardiff , CF10 3AT , UK .
| |
Collapse
|
12
|
Ur Rahman A, Khan S, Khan M. Transport of trans-activator of transcription (TAT) peptide in tumour tissue model: evaluation of factors affecting the transport of TAT evidenced by flow cytometry. ACTA ACUST UNITED AC 2019; 72:519-530. [PMID: 31868235 DOI: 10.1111/jphp.13221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/29/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Trans-activator of transcription (TAT), a cell penetrating peptide, has been explored to overcome resistance to penetration and transport inside the cell, therefore, suggested to be used as drug delivery vector into drug-resistant tumours. The generosity of this study was to evaluate modifiable factors (concentration, temperature, incubation time and spheroid age) on the penetration of TAT. METHODS Multicellular tumour spheroids (MCTS) used as tumour tissue models to mimic some characteristics with in-vivo tumors. Cell monolayer and 3-, 5-, 7-day-old MCTS were incubated with TAT and effects of modifiable factors were determined quantitatively through flow cytometry, based on TAT-positive cell count (%) and mean fluorescence intensity. KEY FINDINGS Enhancing TAT concentration (1, 5 and 25 µm), transport significantly increased (ANOVA, P < 0.0001) in cell monolayer and spheroids. However, rising temperature from 7 to 37°C (t, P > 0.05) and increasing incubation time; 20 min, 1 h and 3 h; (ANOVA, P > 0.05) were statistically non-significant. Moreover, TAT penetration declines as spheroids get older (ANOVA, P < 0.01). CONCLUSION While exploiting MCTS as tumour tissue model, older spheroids could be preferred to target penetration-resistant cells and mimic the in-vivo microenvironment.
Collapse
Affiliation(s)
- Aziz Ur Rahman
- Manchester Pharmacy School, The University of Manchester, Manchester, UK.,Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Shahzeb Khan
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Munasib Khan
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| |
Collapse
|
13
|
Mathieu E, Sipos A, Demeyere E, Phipps D, Sakaveli D, Borbas KE. Lanthanide-based tools for the investigation of cellular environments. Chem Commun (Camb) 2018; 54:10021-10035. [PMID: 30101249 DOI: 10.1039/c8cc05271a] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biological probes constructed from lanthanides can provide a variety of readout signals, such as the luminescence of Eu(iii), Tb(iii), Yb(iii), Sm(iii) and Dy(iii), and the proton relaxation enhancement of Gd(iii) and Eu(ii). For numerous applications the intracellular delivery of the lanthanide probe is essential. Here, we review the methods for the intracellular delivery of non-targeted complexes (i.e. where the overall complex structure enhances cellular uptake), as well as complexes attached to a targeting unit (i.e. to a peptide or a small molecule) that facilitates delivery. The cellular applications of lanthanide-based supramolecules (dendrimers, metal organic frameworks) are covered briefly. Throughout, we emphasize the techniques that can confirm the intracellular localization of the lanthanides and those that enable the determination of the fate of the probes once inside the cell. Finally, we highlight methods that have not yet been applied in the context of lanthanide-based probes, but have been successful in the intracellular delivery of other metal-based probes.
Collapse
Affiliation(s)
- Emilie Mathieu
- Department of Chemistry, Ångström Laboratory, Uppsala University, Box 523, 75120, Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
14
|
Langdon-Jones EE, Jones AB, Williams CF, Hayes AJ, Lloyd D, Mottram HJ, Pope SJA. Anticancer, Azonafide-Inspired Fluorescent Ligands and Their Rhenium(I) Complexes for Cellular Imaging. Eur J Inorg Chem 2017. [DOI: 10.1002/ejic.201601271] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
| | - Ariana B. Jones
- School of Chemistry; Cardiff University; CF10 3AT Cardiff UK
| | - Catrin F. Williams
- School of Engineering; Cardiff University; CF24 3AA Cardiff UK
- School of Biosciences; Cardiff University; CF10 3AT Cardiff UK
| | | | - David Lloyd
- School of Biosciences; Cardiff University; CF10 3AT Cardiff UK
| | - Huw J. Mottram
- School of Pharmacy; Cardiff University; CF10 3NB Cardiff UK
| | | |
Collapse
|
15
|
Shimura M, Szyrwiel L, Matsuyama S, Yamauchi K. Visualization of Intracellular Elements Using Scanning X-Ray Fluorescence Microscopy. Metallomics 2017. [DOI: 10.1007/978-4-431-56463-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
16
|
Wedding JL, Harris HH, Bader CA, Plush SE, Mak R, Massi M, Brooks DA, Lai B, Vogt S, Werrett MV, Simpson PV, Skelton BW, Stagni S. Intracellular distribution and stability of a luminescent rhenium(i) tricarbonyl tetrazolato complex using epifluorescence microscopy in conjunction with X-ray fluorescence imaging. Metallomics 2017; 9:382-390. [DOI: 10.1039/c6mt00243a] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Ranasinghe K, Handunnetti S, Perera IC, Perera T. Synthesis and characterization of novel rhenium(I) complexes towards potential biological imaging applications. Chem Cent J 2016; 10:71. [PMID: 27942267 PMCID: PMC5123207 DOI: 10.1186/s13065-016-0218-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 11/10/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Re(I) tricarbonyl complexes exhibit immense potential as fluorescence imaging agents. However, only a handful of rhenium complexes have been utilized in biological imaging. The present study describes the synthesis of four novel rhenium complexes, their characterization and preliminary biological studies to assess their potential as biological imaging agents. RESULTS Four facial rhenium tricarbonyl complexes containing a pyridyl triazine core, (L1 = 5,5'(3-(2-pyridyl)-1,2,4-triazine-5,6-diyl)-bis-2-furansulfonic acid disodium salt and L2 = (3-(2- pyridyl)-5,6-diphenyl-1,2,4-triazine-4',4''-disulfonic acid sodium salt) have been synthesized by utililzing two different Re metal precursors, Re(CO)5Br and [Re(CO)3(H2O)3]OTf in an organic solvent mixture and water, respectively. The rhenium complexes [Re(CO)3(H2O)L1]+ (1), Re(CO)3L1Br (2), [Re(CO)3(H2O)L2]+ (3), and Re(CO)3L2Br (4), were obtained in 70-85% yield and characterized by 1H NMR, IR, UV, and luminescence spectroscopy. In both H2O and acetonitrile, complexes display a weak absorption band in the visible region which can be assigned to a metal to ligand charge transfer excitation and fluorescent emission lying in the 650-710 nm range. Cytotoxicity assays of complexes 1, 3, and 4 were carried out for rat peritoneal cells. Both plant cells (Allium cepa bulb cells) and rat peritoneal cells were stained using the maximum non-toxic concentration levels of the compounds, 20.00 mg ml-1 for 1 and 3 and 5.00 mg ml-1 for 4 to observe under the epifluorescence microscope. In both cell lines, compound concentrated specifically in the nuclei region. Hence, nuclei showed red fluorescence upon excitation at 550 nm. CONCLUSIONS Four novel rhenium complexes have been synthesized and characterized. Remarkable enhancement of fluorescence upon binding with cells and visible range excitability demonstrates the possibility of using the new complexes in biological applications.Graphical abstractMicrograph of rat peritoneal cells incubated with novel rhenium complex under epifluorescence microscope.
Collapse
Affiliation(s)
- Kokila Ranasinghe
- Department of Chemistry, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Shiroma Handunnetti
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo, Sri Lanka
| | - Inoka C. Perera
- Department of Zoology and Environmental Sciences, University of Colombo, Colombo, Sri Lanka
| | - Theshini Perera
- Department of Chemistry, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| |
Collapse
|
18
|
Direct Correlation Between Zeta Potential and Cellular Uptake of Poly(methacrylic acid) Post‐Modified with Guanidinium Functionalities. MACROMOL CHEM PHYS 2016. [DOI: 10.1002/macp.201600161] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
19
|
GUO ZHENGRONG, PENG HUANYAN, KANG JIWEN, SUN DIANXING. Cell-penetrating peptides: Possible transduction mechanisms and therapeutic applications. Biomed Rep 2016; 4:528-534. [PMID: 27123243 PMCID: PMC4840506 DOI: 10.3892/br.2016.639] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/09/2016] [Indexed: 01/09/2023] Open
Abstract
Cell-penetrating peptides (CPPs), also known as protein transduction domains, are a class of diverse peptides with 5-30 amino acids. CPPs are divided into cationic, amphipathic and hydrophobic CPPs. They are able to carry small molecules, plasmid DNA, small interfering RNA, proteins, viruses, imaging agents and other various nanoparticles across the cellular membrane, resulting in internalization of the intact cargos. However, the mechanisms of CPP internalization remain to be elucidated. Recently, CPPs have received considerable attention due to their high transduction efficiency and low cytotoxicity. These peptides have a significant potential for diagnostic and therapeutic applications, such as delivery of fluorescent or radioactive compounds for imaging, delivery of peptides and proteins for therapeutic application, and delivery of molecules into induced pluripotent stem cells for directing differentiation. The present study reviews the classifications and transduction mechanisms of CPPs, as well as their potential applications.
Collapse
Affiliation(s)
- ZHENGRONG GUO
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
- Department of Gastroenterology, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - HUANYAN PENG
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
- Hebei Medical University Graduate School, Shijiazhuang, Hebei 050017, P.R. China
| | - JIWEN KANG
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
| | - DIANXING SUN
- The Liver Diseases Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, Hebei 050082, P.R. China
| |
Collapse
|
20
|
Abstract
During the three decades of cell-penetrating peptides era the superfamily of CPPs has rapidly expanded, and the quest for new sequences continues. CPPs have been well recognized by scientific community and they have been used for transduction of a wide variety of molecules and particles into cultured cells and in vivo. In parallel with application of CPPs for delivering of active payloads, the mechanisms that such peptides take advantage of for gaining access to cells' insides have been in the focus of intense studies. Although the common denominator "cell penetration" unites all CPPs, the interaction partners on the cell surface, evoked cellular responses and even the uptake mechanisms might greatly vary between different peptide types. Here we present some possibilities for classification of CPPs based on their type of origin, physical-chemical properties, and the extent of modifications and design efforts. We also briefly analyze the internalization mechanisms with regard to their classification into groups based on physical-chemical characteristics.
Collapse
|
21
|
Lee JH, Zhang A, You SS, Lieber CM. Spontaneous Internalization of Cell Penetrating Peptide-Modified Nanowires into Primary Neurons. NANO LETTERS 2016; 16:1509-13. [PMID: 26745653 DOI: 10.1021/acs.nanolett.6b00020] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Semiconductor nanowire (NW) devices that can address intracellular electrophysiological events with high sensitivity and spatial resolution are emerging as key tools in nanobioelectronics. Intracellular delivery of NWs without compromising cellular integrity and metabolic activity has, however, proven difficult without external mechanical forces or electrical pulses. Here, we introduce a biomimetic approach in which a cell penetrating peptide, the trans-activating transcriptional activator (TAT) from human immunodeficiency virus 1, is linked to the surface of Si NWs to facilitate spontaneous internalization of NWs into primary neuronal cells. Confocal microscopy imaging studies at fixed time points demonstrate that TAT-conjugated NWs (TAT-NWs) are fully internalized into mouse hippocampal neurons, and quantitative image analyses reveal an ca. 15% internalization efficiency. In addition, live cell dynamic imaging of NW internalization shows that NW penetration begins within 10-20 min after binding to the membrane and that NWs become fully internalized within 30-40 min. The generality of cell penetrating peptide modification method is further demonstrated by internalization of TAT-NWs into primary dorsal root ganglion (DRG) neurons.
Collapse
Affiliation(s)
- Jae-Hyun Lee
- Department of Chemistry and Chemical Biology and ‡John A. Paulson School of Engineering and Applied Science, Harvard University , Cambridge, Massachusetts 02138, United States
| | - Anqi Zhang
- Department of Chemistry and Chemical Biology and ‡John A. Paulson School of Engineering and Applied Science, Harvard University , Cambridge, Massachusetts 02138, United States
| | - Siheng Sean You
- Department of Chemistry and Chemical Biology and ‡John A. Paulson School of Engineering and Applied Science, Harvard University , Cambridge, Massachusetts 02138, United States
| | - Charles M Lieber
- Department of Chemistry and Chemical Biology and ‡John A. Paulson School of Engineering and Applied Science, Harvard University , Cambridge, Massachusetts 02138, United States
| |
Collapse
|
22
|
Abstract
Cell penetrating peptides (CPP), also known as protein transduction domains (PTD), are small peptides able to carry peptides, proteins, nucleic acid, and nanoparticles, including viral particles, across the cellular membranes into cells, resulting in internalization of the intact cargo. In general, CPPs can be broadly classified into tissue-specific and non-tissue specific peptides, with the latter further sub-divided into three types: (1) cationic peptides of 6-12 amino acids in length comprised predominantly of arginine, lysine and/or ornithine residues; (2) hydrophobic peptides such as leader sequences of secreted growth factors or cytokines; and (3) amphipathic peptides obtained by linking hydrophobic peptides to nuclear localizing signals. Tissue-specific peptides are usually identified by screening of large peptide phage display libraries. These transduction peptides have the potential for a myriad of diagnostic as well as therapeutic applications, ranging from delivery of fluorescent or radioactive compounds for imaging, to delivery of peptides and proteins of therapeutic potential, and improving uptake of DNA, RNA, siRNA and even viral particles. Here we review the potential applications as well as hurdles to the tremendous potential of these CPPs, in particular the cell-type specific peptides.
Collapse
|
23
|
Szyrwiel Ł, Shimura M, Shirataki J, Matsuyama S, Matsunaga A, Setner B, Szczukowski Ł, Szewczuk Z, Yamauchi K, Malinka W, Chavatte L, Łobinski R. A novel branched TAT(47-57) peptide for selective Ni(2+) introduction into the human fibrosarcoma cell nucleus. Metallomics 2015; 7:1155-62. [PMID: 25927891 DOI: 10.1039/c5mt00021a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A TAT47-57 peptide was modified on the N-terminus by elongation with a 2,3-diaminopropionic acid residue and then by coupling of two histidine residues on its N-atoms. This branched peptide could bind to Ni under physiological conditions as a 1 : 1 complex. We demonstrated that the complex was quantitatively taken up by human fibrosarcoma cells, in contrast to Ni(2+) ions. Ni localization (especially at the nuclei) was confirmed by imaging using both scanning X-ray fluorescence microscopy and Newport Green fluorescence. A competitive assay with Newport Green showed that the latter displaced the peptide ligand from the Ni-complex. Ni(2+) delivered as a complex with the designed peptide induced substantially more DNA damage than when introduced as a free ion. The availability of such a construct opens up the way to investigate the importance of the nucleus as a target for the cytotoxicity, genotoxicity or carcinogenicity of Ni(2+).
Collapse
Affiliation(s)
- Łukasz Szyrwiel
- CNRS/UPPA, LCABIE, UMR5254, Hélioparc, 2, av. Pr. Angot, F-64053 Pau, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhang R, Yang XZ, Wang JW, Fu LY, Zhao J, Wang FJ. Evaluating the translocation properties of a new nuclear targeted penetrating peptide using two fluorescent markers. J Drug Target 2015; 23:444-52. [DOI: 10.3109/1061186x.2014.1003068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
25
|
Rizzuti M, Nizzardo M, Zanetta C, Ramirez A, Corti S. Therapeutic applications of the cell-penetrating HIV-1 Tat peptide. Drug Discov Today 2014; 20:76-85. [PMID: 25277319 DOI: 10.1016/j.drudis.2014.09.017] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/11/2014] [Accepted: 09/23/2014] [Indexed: 12/29/2022]
Abstract
Over the past decades, many new therapeutic approaches have been developed for several conditions, including neurodegenerative diseases. However, efficient biodistribution and delivery at biological target sites are hampered by the presence of cell and tissue barriers, and a clinical therapy is prevented by the requirement of invasive administration routes. Candidate drug conjugation to cell-penetrating peptides, which are able to cross cellular membranes and reach biological targets even when administered systemically, represents a promising tool to overcome this issue. Here, we review the biology, classification and mechanisms of internalization of cell-penetrating peptides. We focus our attention on the cell-penetrating peptide: HIV-derived Tat peptide, and discuss its efficient but controversial use in basic, preclinical and clinical research from its discovery to the present day.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Monica Nizzardo
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy.
| | - Chiara Zanetta
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Agnese Ramirez
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| |
Collapse
|
26
|
Sista P, Ghosh K, Martinez JS, Rocha RC. Metallo-Biopolymers: Conjugation Strategies and Applications. POLYM REV 2014. [DOI: 10.1080/15583724.2014.913063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
27
|
Langdon-Jones EE, Symonds NO, Yates SE, Hayes AJ, Lloyd D, Williams R, Coles SJ, Horton PN, Pope SJ. Fluorescent Rhenium-Naphthalimide Conjugates as Cellular Imaging Agents. Inorg Chem 2014; 53:3788-97. [DOI: 10.1021/ic500142z] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Emily E. Langdon-Jones
- School
of Chemistry, Main Building, Cardiff University, Cardiff CF10 3AT, Cymru/Wales, U.K
| | - Nadine O. Symonds
- School
of Chemistry, Main Building, Cardiff University, Cardiff CF10 3AT, Cymru/Wales, U.K
| | - Sara E. Yates
- School
of Chemistry, Main Building, Cardiff University, Cardiff CF10 3AT, Cymru/Wales, U.K
| | - Anthony J. Hayes
- School
of Biosciences, Main Building, Cardiff University, Cardiff CF10 3AT, Cymru/Wales, U.K
| | - David Lloyd
- School
of Biosciences, Main Building, Cardiff University, Cardiff CF10 3AT, Cymru/Wales, U.K
| | - Rebecca Williams
- School
of Biosciences, Main Building, Cardiff University, Cardiff CF10 3AT, Cymru/Wales, U.K
| | - Simon J. Coles
- National
Crystallographic Service, Chemistry, Faculty of Natural and Environmental
Sciences, University of Southampton, Highfield, Southampton, SO17 1BJ, England, U.K
| | - Peter N. Horton
- National
Crystallographic Service, Chemistry, Faculty of Natural and Environmental
Sciences, University of Southampton, Highfield, Southampton, SO17 1BJ, England, U.K
| | - Simon J.A. Pope
- School
of Chemistry, Main Building, Cardiff University, Cardiff CF10 3AT, Cymru/Wales, U.K
| |
Collapse
|
28
|
Hillyar CRT, Cornelissen B, Vallis KA. Uptake, internalization and nuclear translocation of radioimmunotherapeutic agents. Ther Deliv 2014; 5:319-35. [PMID: 24592956 DOI: 10.4155/tde.14.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
Radioimmunotherapy (RIT) agents that incorporate short-range particle-emitting radionuclides exploit the high linear energy transfer of α-particles and Auger electrons. Both are densely ionizing, generate complex DNA double-strand breaks and so are profoundly cytotoxic. Internalizing RIT agents enter tumor cells through receptor-mediated endocytosis and by incorporation of cell-penetrating peptides. Once internalized, some RIT agents mediate escape from endosomes and/or translocate to the nucleus. In the classical nuclear import pathway, α/β-importins recognize nuclear localization sequences in RIT agents. Translocation through nuclear pores enables RIT agents to bind to nuclear targets induced by, for example, cellular stress, growth factors or anticancer therapy, such as γH2AX or p27(KIP-1). This review discusses RIT agents designed to exploit the mechanisms underlying these complex processes and compares them with noninternalizing RIT agents.
Collapse
Affiliation(s)
- Christopher R T Hillyar
- Cancer Research UK/Medical Research Council Gray Institute for Radiation Oncology & Biology, Department of Oncology, University of Oxford, OX3 7DQ, UK
| | | | | |
Collapse
|
29
|
Fischer G, Pan B, Vilceanu D, Hogan QH, Yu H. Sustained relief of neuropathic pain by AAV-targeted expression of CBD3 peptide in rat dorsal root ganglion. Gene Ther 2013; 21:44-51. [PMID: 24152582 PMCID: PMC3881029 DOI: 10.1038/gt.2013.56] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/09/2013] [Indexed: 01/06/2023]
Abstract
The Ca2+ channel-binding domain 3 (CBD3) peptide, derived from the collapsin response mediator protein 2 (CRMP-2), is a recently discovered voltage-gated Ca2+ channel (VGCC) blocker with a preference for CaV2.2. Rodent administration of CBD3 conjugated to cell penetrating motif TAT (TAT-CBD3) has been shown to reduce pain behavior in inflammatory and neuropathic pain models. However, TAT-CBD3 analgesia has limitations, including short half-life, lack of cellular specificity and undesired potential off-site effects. We hypothesized that these issues could be addressed by expressing CBD3 encoded by high-expression vectors in primary sensory neurons. We constructed an adeno-associated viral (AAV) vector expressing recombinant fluorescent CBD3 peptide and injected it into lumbar dorsal root ganglia (DRGs) of rats before spared nerve injury (SNI). We show that selective expression of enhanced green fluorescent protein (EGFP)-CBD3 in lumbar 4 (L4) and L5 DRG neurons and their axonal projections results in effective attenuation of nerve injury-induced neuropathic pain in the SNI model. We conclude that AAV-encoded CBD3 delivered to peripheral sensory neurons through DRG injection may be a valuable approach for exploring the role of presynaptic VGCCs and long-term modulation of neurotransmission, and may also be considered for development as a gene therapy strategy to treat chronic neuropathic pain.
Collapse
Affiliation(s)
- G Fischer
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - B Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - D Vilceanu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Q H Hogan
- 1] Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA [2] Department of Anesthesiology, Medical College of Wisconsin, Zablocki VA Medical Center, Milwaukee, WI, USA
| | - H Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
30
|
Kim Y, Binauld S, Stenzel MH. Zwitterionic guanidine-based oligomers mimicking cell-penetrating peptides as a nontoxic alternative to cationic polymers to enhance the cellular uptake of micelles. Biomacromolecules 2012; 13:3418-26. [PMID: 22946476 DOI: 10.1021/bm301351e] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The aim of this work is to generate polymer micelles decorated with a synthetic version of cell-penetrating peptides, which are often rich in arginine with its positively charged guanidine group. A methacrylate-based monomer with guanidinium as functional groups was prepared using arginine (M-Arg) as a building block, resulting in a zwitterionic monomer. RAFT (reversible addition-fragmentation chain transfer) polymerization was employed to generate triblock copolymers with poly(methyl methacrylate)-block-poly(polyethylene glycol methyl ether methacrylate) as the first two blocks, which were subsequently chain extended with the guanidine-based monomer to generate micelles with guanidinium functional groups on the surface. To simulate the actual oligoarginine peptide, which only carries cationic charges, the carboxylate group of P(M-Arg) was methylated to convert the zwitterionic polymer into a cationic polymer P(Me-M-Arg). For comparison, micelles based on triblock copolymers with a third block with permanently cationic charges, poly(2-methacryolyloxy ethyl) trimethyl ammonium chloride (PTMA), was prepared. The hydrodynamic diameters of the micelles were approximately 30-40 nm based on DLS and TEM. A direct correlation between surface charge (zeta potential ζ) and cytotoxicity was observed. The micelles based on the zwitterionic P(M-Arg) were nontoxic (ζ = -10 mV at pH = 7), while the methylated version P(Me-M-Arg) with a high cationic charge (ζ = +35 mV at pH = 7) were observed to be toxic. The cellular uptake of the block copolymers by OVCAR-3 ovarian cancer cell lines was found to be relatively fast (about 35% in 3 min) reaching an equilibrium after approximately 30 min. Both micelles, with either P(M-Arg) or P(Me-M-Arg) on the surface, showed an enhanced uptake compared to micelles with P(PEGMEMA) as shell only. In fact, the percentage of uptake was similar, with the difference that cells incubated with micelles with P(M-Arg) (zwitterionic) stayed alive, while P(Me-M-Arg) (cationic) led to significant cell death.
Collapse
Affiliation(s)
- Yoseop Kim
- University of New South Wales, Sydney, Australia
| | | | | |
Collapse
|
31
|
Wilson SM, Schmutzler BS, Brittain JM, Dustrude ET, Ripsch MS, Pellman JJ, Yeum TS, Hurley JH, Hingtgen CM, White FA, Khanna R. Inhibition of transmitter release and attenuation of anti-retroviral-associated and tibial nerve injury-related painful peripheral neuropathy by novel synthetic Ca2+ channel peptides. J Biol Chem 2012; 287:35065-35077. [PMID: 22891239 DOI: 10.1074/jbc.m112.378695] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
N-type Ca(2+) channels (CaV2.2) are a nidus for neurotransmitter release and nociceptive transmission. However, the use of CaV2.2 blockers in pain therapeutics is limited by side effects resulting from inhibition of the physiological functions of CaV2.2 within the CNS. We identified an anti-nociceptive peptide (Brittain, J. M., Duarte, D. B., Wilson, S. M., Zhu, W., Ballard, C., Johnson, P. L., Liu, N., Xiong, W., Ripsch, M. S., Wang, Y., Fehrenbacher, J. C., Fitz, S. D., Khanna, M., Park, C. K., Schmutzler, B. S., Cheon, B. M., Due, M. R., Brustovetsky, T., Ashpole, N. M., Hudmon, A., Meroueh, S. O., Hingtgen, C. M., Brustovetsky, N., Ji, R. R., Hurley, J. H., Jin, X., Shekhar, A., Xu, X. M., Oxford, G. S., Vasko, M. R., White, F. A., and Khanna, R. (2011) Suppression of inflammatory and neuropathic pain by uncoupling CRMP2 from the presynaptic Ca(2+) channel complex. Nat. Med. 17, 822-829) derived from the axonal collapsin response mediator protein 2 (CRMP2), a protein known to bind and enhance CaV2.2 activity. Using a peptide tiling array, we identified novel peptides within the first intracellular loop (CaV2.2(388-402), "L1") and the distal C terminus (CaV1.2(2014-2028) "Ct-dis") that bound CRMP2. Microscale thermophoresis demonstrated micromolar and nanomolar binding affinities between recombinant CRMP2 and synthetic L1 and Ct-dis peptides, respectively. Co-immunoprecipitation experiments showed that CRMP2 association with CaV2.2 was inhibited by L1 and Ct-dis peptides. L1 and Ct-dis, rendered cell-penetrant by fusion with the protein transduction domain of the human immunodeficiency virus TAT protein, were tested in in vitro and in vivo experiments. Depolarization-induced calcium influx in dorsal root ganglion (DRG) neurons was inhibited by both peptides. Ct-dis, but not L1, peptide inhibited depolarization-stimulated release of the neuropeptide transmitter calcitonin gene-related peptide in mouse DRG neurons. Similar results were obtained in DRGs from mice with a heterozygous mutation of Nf1 linked to neurofibromatosis type 1. Ct-dis peptide, administered intraperitoneally, exhibited antinociception in a zalcitabine (2'-3'-dideoxycytidine) model of AIDS therapy-induced and tibial nerve injury-related peripheral neuropathy. This study suggests that CaV peptides, by perturbing interactions with the neuromodulator CRMP2, contribute to suppression of neuronal hypersensitivity and nociception.
Collapse
Affiliation(s)
- Sarah M Wilson
- Department of Program in Medical Neurosciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Brian S Schmutzler
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Joel M Brittain
- Department of Program in Medical Neurosciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Erik T Dustrude
- Department of Program in Medical Neurosciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Matthew S Ripsch
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Jessica J Pellman
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Tae-Sung Yeum
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Joyce H Hurley
- Department of Program in Medical Neurosciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Cynthia M Hingtgen
- Department of Program in Medical Neurosciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Fletcher A White
- Department of Program in Medical Neurosciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Anesthesia, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Rajesh Khanna
- Department of Program in Medical Neurosciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202; Sophia Therapeutics LLC, Indianapolis, Indiana 46202.
| |
Collapse
|
32
|
Exploring transduction mechanisms of protein transduction domains (PTDs) in living cells utilizing single-quantum dot tracking (SQT) technology. SENSORS 2012; 12:549-72. [PMID: 22368485 PMCID: PMC3279229 DOI: 10.3390/s120100549] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 12/30/2011] [Accepted: 12/31/2011] [Indexed: 12/04/2022]
Abstract
Specific protein domains known as protein transduction domains (PTDs) can permeate cell membranes and deliver proteins or bioactive materials into living cells. Various approaches have been applied for improving their transduction efficacy. It is, therefore, crucial to clarify the entry mechanisms and to identify the rate-limiting steps. Because of technical limitations for imaging PTD behavior on cells with conventional fluorescent-dyes, how PTDs enter the cells has been a topic of much debate. Utilizing quantum dots (QDs), we recently tracked the behavior of PTD that was derived from HIV-1 Tat (TatP) in living cells at the single-molecule level with 7-nm special precision. In this review article, we initially summarize the controversy on TatP entry mechanisms; thereafter, we will focus on our recent findings on single-TatP-QD tracking (SQT), to identify the major sequential steps of intracellular delivery in living cells and to discuss how SQT can easily provide direct information on TatP entry mechanisms. As a primer for SQT study, we also discuss the latest findings on single particle tracking of various molecules on the plasma membrane. Finally, we discuss the problems of QDs and the challenges for the future in utilizing currently available QD probes for SQT. In conclusion, direct identification of the rate-limiting steps of PTD entry with SQT should dramatically improve the methods for enhancing transduction efficiency.
Collapse
|
33
|
Mäger I, Langel K, Lehto T, Eiríksdóttir E, Langel U. The role of endocytosis on the uptake kinetics of luciferin-conjugated cell-penetrating peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:502-11. [PMID: 22155257 DOI: 10.1016/j.bbamem.2011.11.020] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 11/09/2011] [Accepted: 11/23/2011] [Indexed: 11/16/2022]
Abstract
Cell-penetrating peptides (CPPs) are short cationic/amphipathic peptides that can be used to deliver a variety of cargos into cells. However, it is still debated which routes CPPs employ to gain access to intracellular compartments. To assess this, most previously conducted studies have relied on information which is gained by using fluorescently labeled CPPs. More relevant information whether the internalized conjugates are biologically available has been gathered using end-point assays with biological readouts. Uptake kinetic studies have shed even more light on the matter because the arbitrary choice of end-point might have profound effect how the results could be interpreted. To elucidate uptake mechanisms of CPPs, here we have used a bioluminescence based assay to measure cytosolic delivery kinetics of luciferin-CPP conjugates in the presence of endocytosis inhibitors. The results suggest that these conjugates are delivered into cytosol mainly via macropinocytosis; clathrin-mediated endocytosis and caveolae/lipid raft dependent endocytosis are involved in a smaller extent. Furthermore, we demonstrate how the involved endocytic routes and internalization kinetic profiles can depend on conjugate concentration in case of certain peptides, but not in case of others. The employed internalization route, however, likely dictates the intracellular fate and subsequent trafficking of internalized ligands, therefore emphasizing the importance of our novel findings for delivery vector development.
Collapse
Affiliation(s)
- Imre Mäger
- Institute of Technology, University of Tartu, Tartu, Estonia.
| | | | | | | | | |
Collapse
|
34
|
Abstract
As our knowledge increases about the diversity in uptake mechanisms displayed by cell-penetrating peptides (CPP), the concept of CPP uptake kinetics becomes increasingly complex. Here, we present three different assays that can be used for studying different kinetic aspects of CPP-mediated delivery: intracellular accumulation and membranolytical effects, intracellular CPP-cargo detachment, and finally a functional readout of a biological action from the delivered cargo. Unlike the traditional end-point measurements that give a static postincubation readout, these assays are all dynamic, real-time, in situ measurements obtained during incubation. A combination of some (or all) of these different assays gives us not only interesting kinetic information about the uptake routes but also provides a simple and valuable methodology for the evaluation of potential drug candidates based on the chemical modification of CPPs by cargo attachment.
Collapse
|
35
|
Bonduelle CV, Gillies ER. Dendritic Guanidines as Efficient Analogues of Cell Penetrating Peptides. Pharmaceuticals (Basel) 2010; 3:636-666. [PMID: 27713272 PMCID: PMC4033973 DOI: 10.3390/ph3030636] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 02/10/2010] [Accepted: 03/09/2010] [Indexed: 01/09/2023] Open
Abstract
The widespread application of cell penetrating agents to clinical therapeutics and imaging agents relies on the ability to prepare them on a large scale and to readily conjugate them to their cargos. Dendritic analogues of cell penetrating peptides, with multiple guanidine groups on their peripheries offer advantages as their high symmetry allows them to be efficiently synthesized, while orthogonal functionalities at their focal points allow them to be conjugated to cargo using simple synthetic methods. Their chemical structures and properties are also highly tunable as their flexibility and the number of guanidine groups can be tuned by altering the dendritic backbone or the linkages to the guanidine groups. This review describes the development of cell-penetrating dendrimers based on several different backbones, their structure-property relationships, and comparisons of their efficacies with those of known cell penetrating peptides. The toxicities of these dendritic guanidines are also reported as well as their application towards the intracellular delivery of biologically significant cargos including proteins and nanoparticles.
Collapse
Affiliation(s)
- Colin V Bonduelle
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, N6A 5B7, Canada
| | - Elizabeth R Gillies
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, N6A 5B7, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, 1151 Richmond St., London, N6A 5B9, Canada.
| |
Collapse
|
36
|
Kersemans V, Cornelissen B. Targeting the Tumour: Cell Penetrating Peptides for Molecular Imaging and Radiotherapy. Pharmaceuticals (Basel) 2010; 3:600-620. [PMID: 27713270 PMCID: PMC4033971 DOI: 10.3390/ph3030600] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2009] [Revised: 02/02/2010] [Accepted: 03/10/2010] [Indexed: 11/16/2022] Open
Abstract
Over the last couple of years, the number of original papers and reviews discussing various applications of cell penetrating peptides (CPPs) has grown exponentially. This is not remarkable since CPPs are capable of transporting the most varying cargo across cell membranes which is one of the biggest problems in drug delivery and targeted therapy. In this review, we focus on the use of CPPs and related peptides for delivery of imaging contrast agents and radionuclides to cells and tissues with the ultimate goal of in vivo molecular imaging and molecular radiotherapy of intracellular and even intranuclear targets.
Collapse
Affiliation(s)
- Veerle Kersemans
- Gray Institute for Radiation Oncology and Biology, University of Oxford/Old Road Campus Research Building, Off Roosevelt Drive, Churchill Hospital, Oxford OX3 7DQ, UK.
| | - Bart Cornelissen
- Gray Institute for Radiation Oncology and Biology, University of Oxford/Old Road Campus Research Building, Off Roosevelt Drive, Churchill Hospital, Oxford OX3 7DQ, UK.
| |
Collapse
|
37
|
Said Hassane F, Saleh AF, Abes R, Gait MJ, Lebleu B. Cell penetrating peptides: overview and applications to the delivery of oligonucleotides. Cell Mol Life Sci 2010; 67:715-26. [PMID: 19898741 PMCID: PMC11115801 DOI: 10.1007/s00018-009-0186-0] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 10/02/2009] [Accepted: 10/15/2009] [Indexed: 01/19/2023]
Abstract
Crossing biological barriers represents a major limitation for clinical applications of biomolecules such as nucleic acids, peptides or proteins. Cell penetrating peptides (CPP), also named protein transduction domains, comprise short and usually basic amino acids-rich peptides originating from proteins able to cross biological barriers, such as the viral Tat protein, or are rationally designed. They have emerged as a new class of non-viral vectors allowing the delivery of various biomolecules across biological barriers from low molecular weight drugs to nanosized particles. Encouraging data with CPP-conjugated oligonucleotides have been obtained both in vitro and in vivo in animal models of diseases such as Duchenne muscular dystrophy. Whether CPP-cargo conjugates enter cells by direct translocation across the plasma membrane or by endocytosis remains controversial. In many instances, however, endosomal escape appears as a major limitation of this new delivery strategy.
Collapse
Affiliation(s)
- F. Said Hassane
- UMR 5235 CNRS, Dynamique des Interactions Membranaires Normales et Pathologiques, Université Montpellier 2, 34095 Montpellier Cedex 5, France
| | - A. F. Saleh
- Laboratory of Molecular Biology, Medical Research Council, Hills Road, Cambridge, CB2 0QH UK
| | - R. Abes
- UMR 5235 CNRS, Dynamique des Interactions Membranaires Normales et Pathologiques, Université Montpellier 2, 34095 Montpellier Cedex 5, France
| | - M. J. Gait
- Laboratory of Molecular Biology, Medical Research Council, Hills Road, Cambridge, CB2 0QH UK
| | - Bernard Lebleu
- UMR 5235 CNRS, Dynamique des Interactions Membranaires Normales et Pathologiques, Université Montpellier 2, 34095 Montpellier Cedex 5, France
| |
Collapse
|
38
|
Abstract
Transition metal complexes offer great potential as diagnostic and therapeutic agents, and a growing number of biological applications have been explored. To be effective, these complexes must reach their intended target inside the cell. Here we review the cellular accumulation of metal complexes, including their uptake, localization, and efflux. Metal complexes are taken up inside cells through various mechanisms, including passive diffusion and entry through organic and metal transporters. Emphasis is placed on the methods used to examine cellular accumulation, to identify the mechanism(s) of uptake, and to monitor possible efflux. Conjugation strategies that have been employed to improve the cellular uptake characteristics of metal complexes are also described.
Collapse
Affiliation(s)
- Cindy A. Puckett
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Russell J. Ernst
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Jacqueline K. Barton
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
39
|
Sivapackiam J, Gammon ST, Harpstrite SE, Sharma V. Targeted chemotherapy in drug-resistant tumors, noninvasive imaging of P-glycoprotein-mediated functional transport in cancer, and emerging role of Pgp in neurodegenerative diseases. Methods Mol Biol 2010; 596:141-81. [PMID: 19949924 DOI: 10.1007/978-1-60761-416-6_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multidrug resistance (MDR) mediated by overexpression of P-glycoprotein (Pgp) is one of the best characterized transporter-mediated barriers to successful chemotherapy in cancer patients and is also a rapidly emerging target in the progression of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Therefore, strategies capable of delivering chemotherapeutic agents into drug-resistant tumors and targeted radiopharmaceuticals acting as ultrasensitive molecular imaging probes for detecting functional Pgp expression in vivo could be expected to play a vital role in systemic biology as personalized medicine gains momentum in the twenty-first century. While targeted therapy could be expected to deliver optimal doses of chemotherapeutic drugs into the desired targets, the interrogation of Pgp-mediated transport activity in vivo via noninvasive imaging techniques (SPECT and PET) would be beneficial in stratification of patient populations likely to benefit from a given therapeutic treatment, thereby assisting management of drug resistance in cancer and treatment of neurodegenerative diseases. Both strategies could play a vital role in advancement of personalized treatments in cancer and neurodegenerative diseases. Via this tutorial, authors make an attempt in outlining these strategies and discuss their strengths and weaknesses.
Collapse
Affiliation(s)
- Jothilingam Sivapackiam
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University Medical School, St. Louis, MO, USA
| | | | | | | |
Collapse
|
40
|
Mäger I, Eiríksdóttir E, Langel K, El Andaloussi S, Langel U. Assessing the uptake kinetics and internalization mechanisms of cell-penetrating peptides using a quenched fluorescence assay. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1798:338-43. [PMID: 19909725 DOI: 10.1016/j.bbamem.2009.11.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 09/25/2009] [Accepted: 11/02/2009] [Indexed: 11/15/2022]
Abstract
Cell-penetrating peptides (CPPs) have shown great potency for cargo delivery both in vitro and in vivo. Different biologically relevant molecules need to be delivered into appropriate cellular compartments in order to be active, for instance certain drugs/molecules, e.g. antisense oligonucleotides, peptides, and cytotoxic agents require delivery into the cytoplasm. Assessing uptake mechanisms of CPPs can help to develop novel and more potent cellular delivery vectors, especially in cases when reaching a specific intracellular target requires involvement of a specific internalization pathway. Here we measure the overall uptake kinetics, with emphasis on cytoplasmic delivery, of three cell-penetrating peptides M918, TP10 and pVec using a quenched fluorescence assay. We show that both the uptake levels and kinetic constants depend on the endocytosis inhibitors used in the experiments. In addition, in some cases only the internalization rate is affected by the endocytosis inhibitors while the total uptake level is not and vice versa, which emphasizes importance of kinetic studies when assessing the uptake mechanisms of CPPs. Also, there seems to be a correlation between lower total cellular uptake and higher first-order rate constants. Furthermore, this may indicate simultaneous involvement of different endocytic pathways with different efficacies in the internalization process, as hypothesized but not shown earlier in an uptake kinetics assay.
Collapse
Affiliation(s)
- Imre Mäger
- University of Tartu, Institute of Technology, Nooruse 1-517, 50411 Tartu, Estonia.
| | | | | | | | | |
Collapse
|
41
|
Single-cell imaging of retinal ganglion cell apoptosis with a cell-penetrating, activatable peptide probe in an in vivo glaucoma model. Proc Natl Acad Sci U S A 2009; 106:9391-6. [PMID: 19458250 DOI: 10.1073/pnas.0812884106] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Molecular imaging probes have potential for in vivo identification of apoptosis and other intracellular processes. TcapQ, a cell-penetrating, near-infrared fluorescent peptide probe designed to be optically silent through intramolecular fluorescence quenching and activated by effector caspases, has been previously described and validated in vitro. Herein, using NMDA-induced apoptosis of retinal ganglion cells (RGCs), representing an in vivo rat model of glaucoma, we assessed the ability of TcapQ to image single-cell apoptosis through effector caspase activity. Following intravitreal injection, intracellular TcapQ activation occurred specifically in RGCs, identified individual apoptotic cells, showed a clear dose-response relationship with NMDA, and colocalized with TUNEL labeling in the retina. There was a significant diminution of probe activation following pretreatment with a specific inhibitor of caspase-3. Stereospecificity was also exhibited by the lack of intracellular fluorescence upon administration of the noncleavable isomer, dTcapQ. TcapQ has potential utility in detecting and monitoring single-cell apoptosis in glaucoma in vivo.
Collapse
|
42
|
Aguilera TA, Olson ES, Timmers MM, Jiang T, Tsien RY. Systemic in vivo distribution of activatable cell penetrating peptides is superior to that of cell penetrating peptides. Integr Biol (Camb) 2009; 1:371-81. [PMID: 20023744 PMCID: PMC2796831 DOI: 10.1039/b904878b] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cell penetrating peptides (CPPs) have been developed as vehicles for payload delivery into cells in culture and in animals. However several biologic features limit their usefulness in living animals. Activatable cell penetrating peptides (ACPPs) are polycationic CPPs whose adsorption and cellular uptake are minimized by a covalently attached polyanionic inhibitory domain. Cleavage of the linker connecting the polyanionic and polycationic domains by specific proteases (tumor associated matrix metalloproteases discussed herein) dissociates the polyanion and enables the cleaved ACPP to enter cells. In contrast to their CPP counterpart, ACPPs are relatively nonadherent and distributed uniformly to normal tissues. While nonaarginine (r(9)) CPP administered intravenously into mice initially bind to the local vasculature and redistribute to the liver, where >90% of the injected dose accumulates 30 min after injection. Regardless of the presence of the polyanionic inhibitory domain, confocal imaging of live tissues reveals that the majority of the ACPP and CPP remain in punctate organelles, presumably endosomes. Therefore further improvements in the efficiency of delivery to the cytosol and nucleus are necessary. In addition to improved target specificity, a major advantage of ACPPs over CPPs for potential clinical applications is reduced toxicity. Systemically administered r(9) CPP causes acute toxicity in mice at a dose 4-fold lower than the MMP cleavable ACPP, a complication not observed with an uncleavable ACPP presumably because the polycationic charge remains masked systemically. These data suggest that ACPPs have greater potential than CPPs for systemic delivery of imaging and therapeutic agents.
Collapse
Affiliation(s)
- Todd A Aguilera
- Department of Pharmacology, University of California at San Diego, La Jolla, CA 92093-0647, USA
| | | | | | | | | |
Collapse
|
43
|
Development of an optimized activatable MMP-14 targeted SPECT imaging probe. Bioorg Med Chem 2008; 17:653-9. [PMID: 19109023 DOI: 10.1016/j.bmc.2008.11.078] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 11/18/2008] [Accepted: 11/23/2008] [Indexed: 11/20/2022]
Abstract
Matrix metalloproteinase-14 (MT1-MMP or MMP-14) is a membrane-associated protease implicated in a variety of tissue remodeling processes and a molecular hallmark of select metastatic cancers. The ability to detect MMP-14 in vivo would be useful in studying its role in pathologic processes and may potentially serve as a guide for the development of targeted molecular therapies. Four MMP-14 specific probes containing a positively charged cell penetrating peptide (CPP) d-arginine octamer (r(8)) linked with a MMP-14 peptide substrate and attenuating sequences with glutamate (8e, 4e) or glutamate-glycine (4eg and 4egg) repeating units were modeled using an AMBER force field method. The probe with 4egg attenuating sequence exhibited the highest CPP/attenuator interaction, predicting minimized cellular uptake until cleaved. The in vitro MMP-14-mediated cleavage studies using the human recombinant MMP-14 catalytic domain revealed an enhanced cleavage rate that directly correlated with the linearity of the embedded peptide substrate sequence. Successful cleavage and uptake of a technetium-99m labeled version of the optimal probe was demonstrated in MMP-14 transfected human breast cancer cells. Two-fold reduction of cellular uptake was found in the presence of a broad spectrum MMP inhibitor. The combination of computational chemistry, parallel synthesis and biochemical screening, therefore, shows promise as a set of tools for developing new radiolabeled probes that are sensitive to protease activity.
Collapse
|
44
|
TAT-conjugated nanoparticles for the CNS delivery of anti-HIV drugs. Biomaterials 2008; 29:4429-38. [PMID: 18760470 DOI: 10.1016/j.biomaterials.2008.08.004] [Citation(s) in RCA: 233] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2008] [Accepted: 08/04/2008] [Indexed: 11/22/2022]
Abstract
We have shown that nanoparticles (NPs) conjugated to trans-activating transcriptor (TAT) peptide bypass the efflux action of P-glycoprotein and increase the transport of the encapsulated ritonavir, a protease inhibitor (PI), across the blood-brain-barrier (BBB) to the central nervous system (CNS). A steady increase in the drug parenchyma/capillary ratio over time without disrupting the BBB integrity suggests that TAT-conjugated NPs are first immobilized in the brain vasculature prior to their transport into parenchyma. Localization of NPs in the brain parenchyma was further confirmed with histological analysis of the brain sections. The brain drug level with conjugated NPs was 800-fold higher than that with drug in solution at two weeks. Drug clearance was seen within four weeks. In conclusion, TAT-conjugated NPs enhanced the CNS bioavailability of the encapsulated PI and maintained therapeutic drug levels in the brain for a sustained period that could be effective in reducing the viral load in the CNS, which acts as a reservoir for the replicating HIV-1 virus.
Collapse
|
45
|
Fischer PM. Cellular uptake mechanisms and potential therapeutic utility of peptidic cell delivery vectors: progress 2001-2006. Med Res Rev 2008; 27:755-95. [PMID: 17019680 DOI: 10.1002/med.20093] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cell delivery vectors (CDVs) are short amphipathic and cationic peptides and peptide derivatives, usually containing multiple lysine and arginine residues. They possess inherent membrane activity and can be conjugated or complexed with large impermeable macromolecules and even microscopic particles to facilitate cell entry. Various mechanisms have been proposed but it is now becoming clear that the main port of entry into cells of such CDV constructs involves adsorptive-mediated endocytosis rather than direct penetration of the plasma membrane. It is still unclear, however, how and to what extent CDV constructs are capable of exiting endosomal compartments and reaching their intended cellular site of action, usually the cytosol or the nucleus. Furthermore, although many CDVs can mediate cellular uptake of their cargo and appear comparatively non-toxic to cells in tissue culture, the utility of CDVs for in vivo applications remains poorly understood. Whatever the mechanisms of cell entry and disposition, the overriding question as far as potential pharmacological application of CDV conjugates is concerned is whether or not a therapeutic margin can be achieved by their administration. Such a margin will only result if the intracellular concentration in the target tissues necessary to elicit the biological effect of the CDV cargo can be achieved at systemic CDV exposure levels that are non-toxic to both target and bystander cells. It is proposed that the focus of CDV research now be shifted from mechanistic in vitro studies with labeled but otherwise unconjugated CDVs to in vivo pharmacological and toxicological studies using CDV-derivatized and other cationized forms of inherently non-permeable macromolecules of true therapeutic interest.
Collapse
Affiliation(s)
- Peter M Fischer
- Centre for Biomolecular Sciences and School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom.
| |
Collapse
|
46
|
Isostructural fluorescent and radioactive probes for monitoring neural stem and progenitor cell transplants. Nucl Med Biol 2008; 35:159-69. [DOI: 10.1016/j.nucmedbio.2007.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 09/25/2007] [Accepted: 11/02/2007] [Indexed: 01/17/2023]
|
47
|
Costantini DL, Hu M, Reilly RM. Update:Peptide Motifs for Insertion of Radiolabeled Biomolecules into Cells and Routing to the Nucleus for Cancer Imaging or Radiotherapeutic Applications. Cancer Biother Radiopharm 2008; 23:3-24. [DOI: 10.1089/cbr.2007.0430] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Danny L. Costantini
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Meiduo Hu
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Raymond M. Reilly
- Leslie Dan Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
48
|
Stewart KM, Horton KL, Kelley SO. Cell-penetrating peptides as delivery vehicles for biology and medicine. Org Biomol Chem 2008; 6:2242-55. [DOI: 10.1039/b719950c] [Citation(s) in RCA: 316] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
Foerg C, Merkle HP. On The Biomedical Promise of Cell Penetrating Peptides: Limits Versus Prospects. J Pharm Sci 2008; 97:144-62. [PMID: 17763452 DOI: 10.1002/jps.21117] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cell membrane poses a substantial hurdle to the use of pharmacologically active biomacromolecules that are not per se actively translocated into cells. An appealing approach to deliver such molecules involves tethering or complexing them with so-called cell penetrating peptides (CPPs) that are able to cross the plasma membrane of mammalian cells. The CPP approach is currently a major avenue in engineering delivery systems that are hoped to mediate the non-invasive import of problematic cargos into cells. The large number of different cargo molecules that have been efficiently delivered by CPPs ranges from small molecules to proteins and even liposomes and particles. With respect to the involved mechanism(s) there is increasing evidence for endocytosis as a major route of entry. Moreover, in terms of intracellular trafficking, current data argues for the transport to acidic early endosomal compartments with cytosolic release mediated via retrograde delivery through the Golgi apparatus and the endoplasmic reticulum. The focus of this review is to revisit the performance of cell penetrating peptides for drug delivery. To this aim we cover both accomplishments and failures and report on new prospects of the CPP approach. Besides a selection of successful case histories of CPPs we also review the limitations of CPP mediated translocation. In particular, we comment on the impact of (i) metabolic degradation, (ii) the cell line and cellular differentiation state dependent uptake of CPPs, as well as (iii) the regulation of their endocytic traffic by Rho-family GTPases. Further on, we aim at the identification of promising niches for CPP application in drug delivery. In this context, as inspired by current literature, we focus on three principal areas: (i) the delivery of antineoplastic agents, (ii) the delivery of CPPs as antimicrobials, and (iii) the potential of CPPs to target inflammatory tissues.
Collapse
Affiliation(s)
- Christina Foerg
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Wolfgang-Pauli-Strasse 10, CH-8093 Zurich, Switzerland
| | | |
Collapse
|
50
|
Chen L, Harrison SD. Cell-penetrating peptides in drug development: enabling intracellular targets. Biochem Soc Trans 2007; 35:821-5. [PMID: 17635156 DOI: 10.1042/bst0350821] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A large body of literature has shown that CPPs (cell-penetrating peptides) are capable of carrying macromolecules across the plasma membrane. CPPs can penetrate a wide variety of tissue types and enable modulation of intracellular targets with molecules that, by themselves, are incapable of penetrating cells. As a result, CPPs are recognized for their potential value in validating intracellular targets that could lead to drug discovery programmes [Dietz and Bahr (2004) Mol. Cell Neurosci. 27, 85-131]. The potential for CPP-drug conjugates to be used as human therapeutic agents has not been extensively explored and there is limited knowledge regarding the characteristics of CPPs which are necessary for drug development. A better understanding of the properties of CPPs relating to in vivo pharmacology, pharmacokinetics, pharmacodynamics and safety will continue to inform and encourage novel drug development efforts using CPPs as therapeutics. Here we will discuss areas of interest for drug development of CPP-conjugated compounds.
Collapse
Affiliation(s)
- L Chen
- KAI Pharmaceuticals Inc., 270 Littlefield Avenue, South San Francisco, CA 94080, USA
| | | |
Collapse
|