1
|
Polderdijk SGI, Limzerwala JF, Spiess C. Plasma membrane damage limits cytoplasmic delivery by conventional cell penetrating peptides. PLoS One 2024; 19:e0305848. [PMID: 39226290 PMCID: PMC11371239 DOI: 10.1371/journal.pone.0305848] [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: 02/21/2024] [Accepted: 06/04/2024] [Indexed: 09/05/2024] Open
Abstract
Intracellular delivery of large molecule cargo via cell penetrating peptides (CPPs) is an inefficient process and despite intense efforts in past decades, improvements in efficiency have been marginal. Utilizing a standardized and comparative analysis of the delivery efficiency of previously described cationic, anionic, and amphiphilic CPPs, we demonstrate that the delivery ceiling is accompanied by irreparable plasma membrane damage that is part of the uptake mechanism. As a consequence, intracellular delivery correlates with cell toxicity and is more efficient for smaller peptides than for large molecule cargo. The delivery of pharmaceutically relevant cargo quantities with acceptable toxicity thus seems hard to achieve with the CPPs tested in our study. Our results suggest that any engineered intracellular delivery system based on conventional cationic or amphiphilic CPPs, or the design principles underlying them, needs to accept low delivery yields due to toxicity limiting efficient cytoplasmic uptake. Novel peptide designs based on detailed study of uptake mechanisms are required to overcome these limitations.
Collapse
Affiliation(s)
| | - Jazeel F. Limzerwala
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, United States of America
| | - Christoph Spiess
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, United States of America
| |
Collapse
|
2
|
Lebek T, Malaguti M, Boezio GL, Zoupi L, Briscoe J, Elfick A, Lowell S. PUFFFIN: an ultra-bright, customisable, single-plasmid system for labelling cell neighbourhoods. EMBO J 2024; 43:4110-4135. [PMID: 38997504 PMCID: PMC11405414 DOI: 10.1038/s44318-024-00154-w] [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: 10/28/2023] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
Cell communication coordinates developmental processes, maintains homeostasis, and contributes to disease. Therefore, understanding the relationship between cells in a shared environment is crucial. Here we introduce Positive Ultra-bright Fluorescent Fusion For Identifying Neighbours (PUFFFIN), a cell neighbour-labelling system based upon secretion and uptake of positively supercharged fluorescent protein s36GFP. We fused s36GFP to mNeonGreen or to a HaloTag, facilitating ultra-bright, sensitive, colour-of-choice labelling. Secretor cells transfer PUFFFIN to neighbours while retaining nuclear mCherry, making identification, isolation, and investigation of live neighbours straightforward. PUFFFIN can be delivered to cells, tissues, or embryos on a customisable single-plasmid construct composed of interchangeable components with the option to incorporate any transgene. This versatility enables the manipulation of cell properties, while simultaneously labelling surrounding cells, in cell culture or in vivo. We use PUFFFIN to ask whether pluripotent cells adjust the pace of differentiation to synchronise with their neighbours during exit from naïve pluripotency. PUFFFIN offers a simple, sensitive, customisable approach to profile non-cell-autonomous responses to natural or induced changes in cell identity or behaviour.
Collapse
Affiliation(s)
- Tamina Lebek
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Mattias Malaguti
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Centre for Engineering Biology, Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH9 3FF, UK
| | | | - Lida Zoupi
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
- Simons Initiative for the Developing Brain, The University of Edinburgh, Edinburgh, EH8 9XD, UK
| | | | - Alistair Elfick
- Institute for Bioengineering, School of Engineering, University of Edinburgh, Edinburgh, EH8 3DW, UK
- UK Centre for Mammalian Synthetic Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Sally Lowell
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
3
|
Jones AA, Snow CD. Porous protein crystals: synthesis and applications. Chem Commun (Camb) 2024; 60:5790-5803. [PMID: 38756076 DOI: 10.1039/d4cc00183d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Large-pore protein crystals (LPCs) are an emerging class of biomaterials. The inherent diversity of proteins translates to a diversity of crystal lattice structures, many of which display large pores and solvent channels. These pores can, in turn, be functionalized via directed evolution and rational redesign based on the known crystal structures. LPCs possess extremely high solvent content, as well as extremely high surface area to volume ratios. Because of these characteristics, LPCs continue to be explored in diverse applications including catalysis, targeted therapeutic delivery, templating of nanostructures, structural biology. This Feature review article will describe several of the existing platforms in detail, with particular focus on LPC synthesis approaches and reported applications.
Collapse
Affiliation(s)
- Alec Arthur Jones
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523-1301, USA.
| | - Christopher D Snow
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523-1301, USA.
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523-1301, USA
| |
Collapse
|
4
|
Giancola JB, Grimm JB, Jun JV, Petri YD, Lavis LD, Raines RT. Evaluation of the Cytosolic Uptake of HaloTag Using a pH-Sensitive Dye. ACS Chem Biol 2024; 19:908-915. [PMID: 38525961 PMCID: PMC11186736 DOI: 10.1021/acschembio.3c00713] [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] [Indexed: 03/26/2024]
Abstract
The efficient cytosolic delivery of proteins is critical for advancing novel therapeutic strategies. Current delivery methods are severely limited by endosomal entrapment, and detection methods lack sophistication in tracking the fate of delivered protein cargo. HaloTag, a commonly used protein in chemical biology and a challenging delivery target, is an exceptional model system for understanding and exploiting cellular delivery. Here, we employed a combinatorial strategy to direct HaloTag to the cytosol. We established the use of Virginia Orange, a pH-sensitive fluorophore, and Janelia Fluor 585, a similar but pH-agnostic fluorophore, in a fluorogenic assay to ascertain protein localization within human cells. Using this assay, we investigated HaloTag delivery upon modification with cell-penetrating peptides, carboxyl group esterification, and cotreatment with an endosomolytic agent. We found efficacious cytosolic entry with two distinct delivery methods. This study expands the toolkit for detecting the cytosolic access of proteins and highlights that multiple intracellular delivery strategies can be used synergistically to effect cytosolic access. Moreover, HaloTag is poised to serve as a platform for the delivery of varied cargo into human cells.
Collapse
Affiliation(s)
- JoLynn B. Giancola
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jonathan B. Grimm
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn VA 20147, United States
| | - Joomyung V. Jun
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Yana D. Petri
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Luke D. Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn VA 20147, United States
| | - Ronald T. Raines
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
5
|
Malaguti M, Lebek T, Blin G, Lowell S. Enabling neighbour labelling: using synthetic biology to explore how cells influence their neighbours. Development 2024; 151:dev201955. [PMID: 38165174 PMCID: PMC10820747 DOI: 10.1242/dev.201955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Cell-cell interactions are central to development, but exploring how a change in any given cell relates to changes in the neighbour of that cell can be technically challenging. Here, we review recent developments in synthetic biology and image analysis that are helping overcome this problem. We highlight the opportunities presented by these advances and discuss opportunities and limitations in applying them to developmental model systems.
Collapse
Affiliation(s)
- Mattias Malaguti
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Tamina Lebek
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Guillaume Blin
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Sally Lowell
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
6
|
Yu Q, Bai L, Jiang X. Disulfide Click Reaction for Stapling of S-terminal Peptides. Angew Chem Int Ed Engl 2023; 62:e202314379. [PMID: 37950389 DOI: 10.1002/anie.202314379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 11/12/2023]
Abstract
A disulfide click strategy is disclosed for stapling to enhance the metabolic stability and cellular permeability of therapeutic peptides. A 17-membered library of stapling reagents with adjustable lengths and angles was established for rapid double/triple click reactions, bridging S-terminal peptides from 3 to 18 amino acid residues to provide 18- to 48-membered macrocyclic peptides under biocompatible conditions. The constrained peptides exhibited enhanced anti-HCT-116 activity with a locked α-helical conformation (IC50 =6.81 μM vs. biological incompetence for acyclic linear peptides), which could be unstapled for rehabilitation of the native peptides under the assistance of tris(2-carboxyethyl)phosphine (TCEP). This protocol assembles linear peptides into cyclic peptides controllably to retain the diverse three-dimensional conformations, enabling their cellular uptake followed by release of the disulfides for peptide delivery.
Collapse
Affiliation(s)
- Qing Yu
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, P. R. China
| | - Leiyang Bai
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, P. R. China
| | - Xuefeng Jiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, P. R. China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, P. R. China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, P. R. China
| |
Collapse
|
7
|
Liu J, Jiang J, He M, Chen J, Huang S, Liu Z, Yao C, Chen HJ, Xie X, Wang J. Nanopore Electroporation Device for DNA Transfection into Various Spreading and Nonadherent Cell Types. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50015-50033. [PMID: 37853502 DOI: 10.1021/acsami.3c10939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Cell transfection plays a crucial role in the study of gene function and regulation of gene expression. The existing gene transfection methods, such as chemical carriers, viruses, electroporation, and microinjection, suffer from limitations, including cell type dependence, reliance on cellular endocytosis, low efficiency, safety concerns, and technical complexity. Nanopore-coupled electroporation offers a promising approach to localizing electric fields for efficient cell membrane perforation and nucleic acid transfection. However, the applicability of nanopore electroporation technology across different cell types lacks a systematic investigation. In this study, we explore the potential of nanopore electroporation for transfecting DNA plasmids into various cell types. Our nanopore electroporation device employs track-etched membranes as the core component. We find that nanopore electroporation efficiently transfects adherent cells, including well-spreading epithelial-like HeLa cells, cardiomyocyte-like HL-1 cells, and dendritic-cell-like DC2.4 cells. However, it shows a limited transfection efficiency in weakly spreading macrophages (RAW264.7) and suspension cells (Jurkat). To gain insights into these observations, we develop a COMSOL model, revealing that nanopore electroporation better localizes the electric field on adherent and well-spreading cells, promoting favorable membrane poration conditions. Our findings provide valuable references for advancing nanopore electroporation as a high-throughput, safe, and efficient gene transfection platform.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510080 Guangzhou, People's Republic of China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Juan Jiang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510080 Guangzhou, People's Republic of China
| | - Mengyi He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Jiayi Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Shuang Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Zhengjie Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Chuanjie Yao
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Xi Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510080 Guangzhou, People's Republic of China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, 510006 Guangzhou, People's Republic of China
| | - Ji Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, 510080 Guangzhou, People's Republic of China
| |
Collapse
|
8
|
Wang C, Zhu Y. A programmable protein delivery tool derived from bacterial syringe. Sci Bull (Beijing) 2023; 68:2125-2127. [PMID: 37625971 DOI: 10.1016/j.scib.2023.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Affiliation(s)
- Changguo Wang
- The MOE Key Laboratory for Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Yongqun Zhu
- The MOE Key Laboratory for Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Department of Gastroenterology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
9
|
Hotta M, Hayase K, Kitanaka A, Li T, Takeoka S. Development of the observation of membrane fusion with label-free liposomes by calcium imaging. Biochem Biophys Rep 2023; 34:101483. [PMID: 37250982 PMCID: PMC10209117 DOI: 10.1016/j.bbrep.2023.101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/31/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Liposomes are artificial vesicles composed of lipid bilayers that have enabled drugs to be encapsulated and delivered to tumor tissue. Membrane-fusogenic liposomes fuse with the plasma membranes of cells to deliver encapsulated drugs directly to the cytosol, which makes it a promising method for rapid and highly efficient drug delivery. In a previous study, liposomal lipid bilayers were labeled with fluorescent probes, and colocalization of labeled lipids with plasma membrane was observed under a microscope. However, there was concern that fluorescent labeling would affect lipid dynamics and cause liposomes to acquire membrane fusogenic ability. In addition, encapsulation of hydrophilic fluorescent substances in the inner aqueous phase sometimes requires an additional step of removing unencapsulated substances after preparation, and there is a risk of leakage. Herein, we propose a new method to observe cell interaction with liposomes without labeling. Our laboratory has developed two types of liposomes with different cellular internalization pathways, i.e., endocytosis and membrane fusion. We found that cytosolic calcium influx would be triggered following the internalization of cationic liposomes, and different cell entry routes led to different calcium responses. Thus, the correlation between cell entry routes and calcium responses could be utilized to study liposome-cell interactions without fluorescent labeling lipids. Briefly, liposomes were added to phorbol 12-myristate 13-acetate (PMA)-primed THP-1 cells, and calcium influx was measured by time-lapse imaging using a fluorescent indicator (Fura 2-AM). Liposomes with high membrane fusogenic ability elicited a strong transient calcium response immediately after adding liposomes, whereas those taken up mainly by endocytosis elicited multiple weak calcium responses. In order to verify the cell entry routes, we also tracked the intracellular distribution of fluorescent-labeled liposomes in PMA-primed THP-1 cells using a confocal laser scanning microscope. It was shown that for fusogenic liposomes, colocalization with plasma membrane occurred at the same time as calcium elevation, whereas for liposomes with a high endocytosis potential, fluorescent dots were observed in the cytoplasm, suggesting the cell internalization by endocytosis. These results suggested that the calcium response patterns correspond to cell entry routes, and membrane fusion can be observed by calcium imaging.
Collapse
Affiliation(s)
- Morihiro Hotta
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Kengo Hayase
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Aya Kitanaka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Tianshu Li
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| |
Collapse
|
10
|
Shin S, Awuah Boadi E, Shah S, Ezell M, Li P, Bandyopadhyay BC. Anti-inflammatory role of extracellular l-arginine through calcium sensing receptor in human renal proximal tubular epithelial (HK-2) cells. Int Immunopharmacol 2023; 117:109853. [PMID: 36827919 PMCID: PMC10124988 DOI: 10.1016/j.intimp.2023.109853] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/24/2023]
Abstract
Renal tubular epithelial cells are capable of synthesizing interleukins (IL) in response to a variety of proinflammatory cytokines. Moreover, elevated urinary levels of IL have been shown in patients with various forms of nephritic diseases. However, the underlying intracellular signaling mechanism is unclear. Here we show the immunological signaling role of l-Arginine (l-Arg) through Ca2+-sensing receptor (CaSR) in human kidney 2 (HK-2) renal proximal tubular epithelial cells, using Ca2+ imaging and patch clamp techniques and its mechanistic link to the downstream cellular function. Both pharmacological and siRNA inhibitors support the activation CaSR by extracellular l-Arg to induced Ca2+ entry via a Transient receptor potential canonical (TRPC) channel in HK-2 cells mainly through the receptor operated Ca2+ entry (ROCE). Activation of CaSR by l-Arg led to the rise in p-p38/p38 expression suggesting [Ca2+]i as a regulator for p38-signaling pathways. Notably, l-Arg activated CaSR-induced Ca2+ signaling reduced the expressions of key fibrotic, inflammatory, and apoptotic genes, suggesting its nephroprotective role via Ca2+ signaling through CaSR in HK-2 cells. Since we found that the IL-6 expressions were inversely proportional to the increasing concentrations of l-Arg in HK-2 cells, we measured the release of IL-6, which steadily decreased as the concentrations of l-Arg were elevated. Taken together, extracellular l-Arg is a negative regulator for IL-6-induced inflammatory process, through the activation of CaSR and TRPC channel by ROCE pathway and can have a potential to alleviate inflammatory renal diseases.
Collapse
Affiliation(s)
- Samuel Shin
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Eugenia Awuah Boadi
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Saloni Shah
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Madison Ezell
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Peijun Li
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, Washington, DC 20037, USA; Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC 20064, USA.
| |
Collapse
|
11
|
Shi J, Wang H, Wang Y, Peng Y, Huang X, Zhang Y, Geng H, Wang Y, Li X, Liu C, Liu C. Mitochondrion-targeting and in situ photocontrolled protein delivery via photocages. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 238:112624. [PMID: 36521315 DOI: 10.1016/j.jphotobiol.2022.112624] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/21/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Defects in mitochondrial proteostasis contribute to many disorders, including cancer, neurodegeneration, and metabolic and genetic diseases. A strategy aimed at restoring the damaged mitochondrial proteostasis is the mitochondrion-targeting and carrier-free delivery of exogenous functional proteins that can replace the endogenous dysfunctional proteins. The modification of a protein with a photolabile protecting group (PPG, i.e., photocage group) can be activated in situ by response to illumination, leading to release of the protein from its photocage. Here, the Cys and peptide photocages with coumarin were first prepared and characterized for proof of concept. Then, we designed a pair of photocage groups PPG-RhB and PPG-TPP using coumarin and mitochondrion-targeting Rhodamine B (RhB) and triphenylphosphine (TPP), and another pair of organelle-nontarget photocage groups Br-PPG and NO2-PPG for comparison. The proteins modified with these two pairs of photocage groups undergo photolysis in solutions, and can penetrate cell membrane toward their destinations in the carrier-free fashions. The intracellular protein photocages are in situ activated by illumination at 405 nm, and the proteins are released from their photocages in mitochondria and cytoplasm, respectively. This strategy of light-responsive and carrier-free cellular delivery enables mitochondrial and cytoplasmic accumulation of exogenous proteins.
Collapse
Affiliation(s)
- Jiayuan Shi
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Huiling Wang
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Yuhui Wang
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Yujie Peng
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Xiaoping Huang
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Yunfeng Zhang
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Hongen Geng
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Yi Wang
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Xiang Li
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China; College of Health Science and Engineering, Hubei University, Wuhan 430062, Hubei, PR China.
| | - Chunrong Liu
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China
| | - Changlin Liu
- Key Laboratory of Pesticide and Chemical Biology of Ministry Education, School of Chemistry, Central China Normal University, Wuhan 430079, Hubei, PR China.
| |
Collapse
|
12
|
Rajapaksha SP, Nawagamuwage SU. Anticorrelated position fluctuation of lipids in forming membrane water pores: molecular dynamics simulations study with dengue virus capsid protein. J Biomol Struct Dyn 2022; 40:11395-11404. [PMID: 34343444 DOI: 10.1080/07391102.2021.1958698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The traffic of molecules into or out of cells is regulated by many membrane-associated mechanisms. Membrane pores are considered as one of the major passage mechanisms, although molecular-level understanding of pore formation is still vague. The opening of a membrane pore depends on many factors, including the influence of some proteins. The ability of the cell-penetrating peptides and supercharged proteins to form membrane pores has been reported. We studied pore formation through dipalmitoylphosphatidylcholine (DPPC) lipid bilayers by supercharged dengue virus capsid (C) protein. Atomistic molecular dynamics simulations confirmed the formation of membrane pores by a combined effect of the C protein and the membrane electric field. Analyses of simulated trajectories showed highly correlated vertical position fluctuations between the Cα atom of the membrane-anchored arginine residues and the phosphorus atoms of the surrounding DPPC lipids. Certain regions of the bilayer were negatively correlated while the others were positively correlated with respect to the fluctuations of the Cα atom of the anchored arginine residues. When positively correlated lipids in one leaflet vertically aligned with the negatively correlated lipids in the other leaflet, a local anticorrelated region was generated by weakening the bilayer. The membrane pore was always formed close to this anticorrelated region. Once formed, the C protein followed the hydrated pathway provided by the water-filled pores to cross the membrane.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Suneth P Rajapaksha
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Sithara U Nawagamuwage
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| |
Collapse
|
13
|
Pistono P, Huang P, Brauer DD, Francis MB. Fitness Landscape-Guided Engineering of Locally Supercharged Virus-like Particles with Enhanced Cell Uptake Properties. ACS Chem Biol 2022; 17:3367-3378. [PMID: 36378277 PMCID: PMC9764284 DOI: 10.1021/acschembio.2c00318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022]
Abstract
Protein-based nanoparticles are useful models for the study of self-assembly and attractive candidates for drug delivery. Virus-like particles (VLPs) are especially promising platforms for expanding the repertoire of therapeutics that can be delivered effectively as they can deliver many copies of a molecule per particle for each delivery event. However, their use is often limited due to poor uptake of VLPs into mammalian cells. In this study, we use the fitness landscape of the bacteriophage MS2 VLP as a guide to engineer capsid variants with positively charged surface residues to enhance their uptake into mammalian cells. By combining mutations with positive fitness scores that were likely to produce assembled capsids, we identified two key double mutants with internalization efficiencies as much as 67-fold higher than that of wtMS2. Internalization of these variants with positively charged surface residues depends on interactions with cell surface sulfated proteoglycans, and yet, they are biophysically similar to wtMS2 with low cytotoxicity and an overall negative charge. Additionally, the best-performing engineered MS2 capsids can deliver a potent anticancer small-molecule therapeutic with efficacy levels similar to antibody-drug conjugates. Through this work, we were able to establish fitness landscape-based engineering as a successful method for designing VLPs with improved cell penetration. These findings suggest that VLPs with positive surface charge could be useful in improving the delivery of small-molecule- and nucleic acid-based therapeutics.
Collapse
Affiliation(s)
- Paige
E. Pistono
- Department
of Chemistry, University of California, Berkeley, California94720, United States
| | - Paul Huang
- Department
of Chemistry, University of California, Berkeley, California94720, United States
| | - Daniel D. Brauer
- Department
of Chemistry, University of California, Berkeley, California94720, United States
| | - Matthew B. Francis
- Department
of Chemistry, University of California, Berkeley, California94720, United States
- Materials
Sciences Division, Lawrence Berkeley National
Laboratory, Berkeley, California94720, United States
| |
Collapse
|
14
|
Wang L, Geng J, Chen L, Guo X, Wang T, Fang Y, Belingon B, Wu J, Li M, Zhan Y, Shang W, Wan Y, Feng X, Li X, Wang H. Improved transfer efficiency of supercharged 36 + GFP protein mediate nucleic acid delivery. Drug Deliv 2022; 29:386-398. [PMID: 35075948 PMCID: PMC8794074 DOI: 10.1080/10717544.2022.2030430] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 12/28/2022] Open
Abstract
The potential of nucleic acid therapeutics to treat diseases by targeting specific cells has resulted in its increasing number of uses in clinical settings. However, the major challenge is to deliver bio-macromolecules into target cells and/or subcellular locations of interest ahead in the development of delivery systems. Although, supercharged residues replaced protein 36 + GFP can facilitate itself and cargoes delivery, its efficiency is still limited. Therefore, we combined our recent progress to further improve 36 + GFP based delivery efficiency. We found that the penetration efficacy of 36 + GFP protein was significantly improved by fusion with CPP-Dot1l or treatment with penetration enhancer dimethyl sulfoxide (DMSO) in vitro. After safely packaged with plasmid DNA, we found that the efficacy of in vitro and in vivo transfection mediated by 36 + GFP-Dot1l fusion protein is also significantly improved than 36 + GFP itself. Our findings illustrated that fusion with CPP-Dot1l or incubation with DMSO is an alternative way to synergically promote 36 + GFP mediated plasmid DNA delivery in vitro and in vivo.
Collapse
Affiliation(s)
- Lidan Wang
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Jingping Geng
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Linlin Chen
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Affiliated Ren He Hospital of China Three Gorges University, Yichang, China
| | - Xiangli Guo
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Tao Wang
- The First Clinical Medical College of China Three Gorges University, Yichang, China
| | - Yanfen Fang
- College of Biological and Pharmaceutical Sciences, China Three Gorges University, Hubei, China
| | - Bonn Belingon
- School of Medicine, Institute of Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jiao Wu
- Affiliated Ren He Hospital of China Three Gorges University, Yichang, China
| | - Manman Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Ying Zhan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Wendou Shang
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Yingying Wan
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Xuemei Feng
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Xianghui Li
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Hu Wang
- Department of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang, China
| |
Collapse
|
15
|
Morris DP, Snipes LC, Hill SA, Woods MM, Mbugua MM, Wade LR, McMurry JL. A reversible cell penetrating peptide-cargo linkage allows dissection of cell penetrating peptide- and cargo-dependent effects on internalization and identifies new functionalities of putative endolytic peptides. Front Pharmacol 2022; 13:1070464. [PMID: 36479201 PMCID: PMC9720253 DOI: 10.3389/fphar.2022.1070464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022] Open
Abstract
Cell penetrating peptides (CPPs) are a promising technology for therapeutic delivery of macromolecular cargos. CPPs have generally used covalent linkages to cargo, ensuring a common fate as one molecule. Conversely, our CPP-adaptor, TAT-CaM, noncovalently binds calmodulin binding sequence (CBS)-containing cargos in calcium rich media then dissociates in the calcium-poor endosomal environment following internalization, enhancing endosomal escape relative to standard CPPs. In this study, we report cell entry of positively charged protein cargos that were not increased by TAT-CaM while cargos based on the negatively charged maltose binding protein (MBP) displayed little intrinsic internalization but were internalized by TAT-CaM. In addition, association of positively charged proteins with negatively charged nucleic acids reduced internalization. This evidence points to the dominant role cargo charge plays in apparent CPP effectiveness. There has been little systematic investigation as to how interaction between CPPs and cargos impacts internalization efficiency. Our adaptors provide a tool that allows combinatorial assays to detect emergent properties. Toward this end we added 4 endolytic peptide (EP) sequences between cargo CBS and MBP moieties to create 4 new cargos and between TAT and CaM to create 4 new adaptors. The new cargos were assayed for internalization alone and with a panel of CPP-adaptors to identify combinations that displayed increased internalization efficiency or other properties. Among the most important results, addition of the EP LAH4 improved adaptor performance and provided some CPP capability to cargos. MBP-LAH4-CBS was internalized more effectively by most adaptors, suggesting this sequence has general stimulatory ability. Two other EPs, Aurein 1.2 and HA2, also provided some CPP capability to their MBP cargos but were unexpectedly antagonistic to internalization by most adaptors due to retention of adaptor/cargo complexes on the cell surface. We thus identified LAH4 as stimulator of internalization in both adaptors and cargos and uncovered new functionality for Aurein 1.2 and HA2, which may be related to their identification as EPs. Future experiments will test new endolytic capabilities made possible with combinatorial approaches.
Collapse
|
16
|
Cavazzini D, Spagnoli G, Mariz FC, Reggiani F, Maggi S, Franceschi V, Donofrio G, Müller M, Bolchi A, Ottonello S. Enhanced immunogenicity of a positively supercharged archaeon thioredoxin scaffold as a cell-penetrating antigen carrier for peptide vaccines. Front Immunol 2022; 13:958123. [PMID: 36032169 PMCID: PMC9405434 DOI: 10.3389/fimmu.2022.958123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
Polycationic resurfaced proteins hold great promise as cell-penetrating bioreagents but their use as carriers for the intracellular delivery of peptide immuno-epitopes has not thus far been explored. Here, we report on the construction and functional characterization of a positively supercharged derivative of Pyrococcus furiosus thioredoxin (PfTrx), a thermally hyperstable protein we have previously validated as a peptide epitope display and immunogenicity enhancing scaffold. Genetic conversion of 13 selected amino acids to lysine residues conferred to PfTrx a net charge of +21 (starting from the -1 charge of the wild-type protein), along with the ability to bind nucleic acids. In its unfused form, +21 PfTrx was readily internalized by HeLa cells and displayed a predominantly cytosolic localization. A different intracellular distribution was observed for a +21 PfTrx-eGFP fusion protein, which although still capable of cell penetration was predominantly localized within endosomes. A mixed cytosolic/endosomal partitioning was observed for a +21 PfTrx derivative harboring three tandemly repeated copies of a previously validated HPV16-L2 (aa 20-38) B-cell epitope grafted to the display site of thioredoxin. Compared to its wild-type counterpart, the positively supercharged antigen induced a faster immune response and displayed an overall superior immunogenicity, including a substantial degree of self-adjuvancy. Altogether, the present data point to +21 PfTrx as a promising novel carrier for intracellular antigen delivery and the construction of potentiated recombinant subunit vaccines.
Collapse
Affiliation(s)
- Davide Cavazzini
- Department of Chemistry, Life Sciences & Environmental Sustainability, University of Parma, Parma, Italy
| | - Gloria Spagnoli
- Department of Chemistry, Life Sciences & Environmental Sustainability, University of Parma, Parma, Italy
| | - Filipe Colaco Mariz
- German Cancer Research Center (DKFZ), Tumorvirus-specific Vaccination Strategies (F035), Heidelberg, Germany
| | - Filippo Reggiani
- Department of Chemistry, Life Sciences & Environmental Sustainability, University of Parma, Parma, Italy
| | - Stefano Maggi
- Department of Chemistry, Life Sciences & Environmental Sustainability, University of Parma, Parma, Italy
| | | | - Gaetano Donofrio
- Department of Veterinary Science, University of Parma, Parma, Italy
- Interdepartmental Center Biopharmanet-Tec, University of Parma, Parma, Italy
| | - Martin Müller
- German Cancer Research Center (DKFZ), Tumorvirus-specific Vaccination Strategies (F035), Heidelberg, Germany
- *Correspondence: Martin Müller, ; Angelo Bolchi,
| | - Angelo Bolchi
- Department of Chemistry, Life Sciences & Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Center Biopharmanet-Tec, University of Parma, Parma, Italy
- *Correspondence: Martin Müller, ; Angelo Bolchi,
| | - Simone Ottonello
- Department of Chemistry, Life Sciences & Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Center Biopharmanet-Tec, University of Parma, Parma, Italy
| |
Collapse
|
17
|
Negi S, Hamori M, Kawahara-Nakagawa Y, Imanishi M, Kurehara M, Kitada C, Kawahito Y, Kishi K, Manabe T, Kawamura N, Kitagishi H, Mashimo M, Shibata N, Sugiura Y. Importance of two-dimensional cation clusters induced by protein folding in intrinsic intracellular membrane permeability. RSC Chem Biol 2022; 3:1076-1084. [PMID: 35975000 PMCID: PMC9347356 DOI: 10.1039/d2cb00098a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
We investigated the cell penetration of Sp1 zinc finger proteins (Sp1 ZF) and the mechanism via which the total cationic charge and distribution of cationic residues on the protein surface affect intracellular trafficking. Sp1 ZFs showed intrinsic cell membrane permeability. The intracellular transfer of Sp1 ZFs other than 1F3 was dependent on the total cationic charge. Investigation of the effect of cationic residue distribution on intracellular membrane permeability revealed that the cellular uptake of unfolded Zn2+-non-coordinating Ala mutants was lower than that of the wild type. Therefore, the total cationic charge and distribution of cationic residues on the protein played crucial roles in intracellular translocation. Mutational studies revealed that the two-dimensional cation cluster on the protein surface significantly improved their cellular uptake. This study will contribute to the design of artificial cargoes that can efficiently transport target substances into cells.
Collapse
Affiliation(s)
- Shigeru Negi
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Mami Hamori
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Yuka Kawahara-Nakagawa
- Graduate School of Life Science, University of Hyogo 3-2-1 Kouto, Kamigori-cho Ako-gun Hyogo 678-1297 Japan
| | - Miki Imanishi
- Institute for Chemical Research, Kyoto University Uji Kyoto 611-0011 Japan
| | - Miku Kurehara
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Chieri Kitada
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Yuri Kawahito
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Kanae Kishi
- Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3 Kasumi Minami-ku Hiroshima 734-8553 Japan
| | - Takayuki Manabe
- Clinical Research Support Center, Asahikawa Medical University Hospital 2-1-1-1 Midorigaokahigashi Asahikawa 078-8510 Japan
| | - Nobuyuki Kawamura
- Education Center for Pharmacy, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences 265-1 Higashijima, Akiha-ku Niigata City Niigata 956-8603 Japan
| | - Hiroaki Kitagishi
- Department of Molecular Chemistry and Biochemistry, Faculty of Science and Engineering, Doshisha University Kyotanabe Kyoto 610-0321 Japan
| | - Masato Mashimo
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Nobuhito Shibata
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Yukio Sugiura
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| |
Collapse
|
18
|
Mandal S, Brik A. Probing the cell delivery of synthetic diubiquitin chains. Chem Commun (Camb) 2022; 58:8782-8785. [PMID: 35837864 PMCID: PMC9350984 DOI: 10.1039/d2cc02476d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, the live-cell delivery of structurally different synthetic diubiquitin chains was examined. We found that the combination of structural variations of the Ub chains (intrinsic factors); nature of CPP and CPP-protein linkage (extrinsic factors) influence their delivery. In this study, the live-cell delivery of structurally different synthetic diubiquitin chains was examined.![]()
Collapse
Affiliation(s)
- Shaswati Mandal
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa, 3200008, Israel.
| | - Ashraf Brik
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa, 3200008, Israel.
| |
Collapse
|
19
|
Mallick AM, Tripathi A, Mishra S, Mukherjee A, Dutta C, Chatterjee A, Sinha Roy R. Emerging Approaches for Enabling RNAi Therapeutics. Chem Asian J 2022; 17:e202200451. [PMID: 35689534 DOI: 10.1002/asia.202200451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/04/2022] [Indexed: 11/07/2022]
Abstract
RNA interference (RNAi) is a primitive evolutionary mechanism developed to escape incorporation of foreign genetic material. siRNA has been instrumental in achieving the therapeutic potential of RNAi by theoretically silencing any gene of interest in a reversible and sequence-specific manner. Extrinsically administered siRNA generally needs a delivery vehicle to span across different physiological barriers and load into the RISC complex in the cytoplasm in its functional form to show its efficacy. This review discusses the designing principles and examples of different classes of delivery vehicles that have proved to be efficient in RNAi therapeutics. We also briefly discuss the role of RNAi therapeutics in genetic and rare diseases, epigenetic modifications, immunomodulation and combination modality to inch closer in creating a personalized therapy for metastatic cancer. At the end, we present, strategies and look into the opportunities to develop efficient delivery vehicles for RNAi which can be translated into clinics.
Collapse
Affiliation(s)
- Argha M Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Archana Tripathi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Sukumar Mishra
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Asmita Mukherjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Chiranjit Dutta
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.,Present address:Department of Biological Sciences, NUS Environmental Research Institute (NERI), National University of Singapore (NUS), Block S2 #05-01, 16 Science Drive 4, Singapore, 117558, Singapore
| | - Ananya Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Rituparna Sinha Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.,Centre for Advanced Functional Materials, Indian Institute of Science Education and Research Kolkata, 741246, Mohanpur, India.,Centre for Climate and Environmental Studies, Indian Institute of Science Education and Research Kolkata, 741246, Mohanpur, India
| |
Collapse
|
20
|
Olivieri PH, Jesus MB, Nader HB, Justo GZ, Sousa AA. Cell-surface glycosaminoglycans regulate the cellular uptake of charged polystyrene nanoparticles. NANOSCALE 2022; 14:7350-7363. [PMID: 35535683 DOI: 10.1039/d1nr07279j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Engineered nanoparticles approaching the cell body will first encounter and interact with cell-surface glycosaminoglycans (GAGs) before reaching the plasma membrane and becoming internalized. However, how surface GAGs may regulate the cellular entry of nanoparticles remains poorly understood. Herein, it is shown that the surface GAGs of Chinese hamster ovary cells perform as a charge-based barrier against the cellular internalization of anionic polystyrene nanoparticles (PS NPs). In contrast, cationic PS NPs interact favorably with the surface GAGs and thereby are efficiently internalized. Anionic PS NPs eventually reaching the plasma membrane bind to scavenger receptors and are endocytosed by clathrin-mediated and lipid raft/cholesterol-dependent mechanisms, whereas cationic PS NPs are primarily internalized via clathrin-mediated endocytosis and macropinocytosis. Upon the enzymatic shedding of surface GAGs, the uptake of anionic PS NPs increases while that of cationic PS NPs is dramatically reduced. Interestingly, the diminished uptake of cationic PS NPs is observed only when heparan sulfate, but not chondroitin sulfate, is cleaved from the cell surface. Heparan sulfate therefore serves as anchors/first receptors to facilitate the cellular entry of cationic PS NPs. These findings contribute to advance the basic science of nanoparticle endocytosis while also having important implications for the use of engineered nanocarriers as intracellular drug-delivery systems.
Collapse
Affiliation(s)
- Paulo H Olivieri
- Department of Biochemistry, Federal University of São Paulo, São Paulo, SP 04044-020, Brazil.
| | - Marcelo B Jesus
- Department of Biochemistry & Tissue Biology, University of Campinas, Campinas, SP 13083-970, Brazil
| | - Helena B Nader
- Department of Biochemistry, Federal University of São Paulo, São Paulo, SP 04044-020, Brazil.
| | - Giselle Z Justo
- Department of Pharmaceutical Sciences and Department of Biochemistry, Federal University of São Paulo, Diadema, SP 09972-270, Brazil.
| | - Alioscka A Sousa
- Department of Biochemistry, Federal University of São Paulo, São Paulo, SP 04044-020, Brazil.
| |
Collapse
|
21
|
Exogenous loading of extracellular vesicles, virus-like particles, and lentiviral vectors with supercharged proteins. Commun Biol 2022; 5:485. [PMID: 35590035 PMCID: PMC9120435 DOI: 10.1038/s42003-022-03440-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 05/03/2022] [Indexed: 12/19/2022] Open
Abstract
Cell membrane-based biovesicles (BVs) are important candidate drug delivery vehicles and comprise extracellular vesicles, virus-like particles, and lentiviral vectors. Here, we introduce a non-enzymatic assembly of purified BVs, supercharged proteins, and plasmid DNA called pDNA-scBVs. This multicomponent vehicle results from the interaction of negative sugar moieties on BVs and supercharged proteins that contain positively charged amino acids on their surface to enhance their affinity for pDNA. pDNA-scBVs were demonstrated to mediate floxed reporter activation in culture by delivering a Cre transgene. We introduced pDNA-scBVs containing both a CRE-encoding plasmid and a BV-packaged floxed reporter into the brains of Ai9 mice. Successful delivery of both payloads by pDNA-scBVs was confirmed with reporter signal in the striatal brain region. Overall, we developed a more efficient method to load isolated BVs with cargo that functionally modified recipient cells. Augmenting the natural properties of BVs opens avenues for adoptive extracellular interventions using therapeutic loaded cargo.
Collapse
|
22
|
Brain Endothelial Cells Utilize Glycolysis for the Maintenance of the Transcellular Permeability. Mol Neurobiol 2022; 59:4315-4333. [PMID: 35508867 DOI: 10.1007/s12035-022-02778-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/19/2022] [Indexed: 10/18/2022]
Abstract
Among the components of the blood-brain barrier (BBB), endothelial cells (ECs) play an important role in supplying limited materials, especially glucose, to the brain. However, the mechanism by which glucose is metabolized in brain ECs is still elusive. To address this topic, we assessed the metabolic signature of glucose utilization using live-cell metabolic assays and liquid chromatography-tandem mass spectrometry metabolomic analysis. We found that brain ECs are highly dependent on aerobic glycolysis, generating lactate as its final product with minimal consumption of glucose. Glucose treatment decreased the oxygen consumption rate in a dose-dependent manner, indicating the Crabtree effect. Moreover, when glycolysis was inhibited, brain ECs showed impaired permeability to molecules utilizing transcellular pathway. In addition, we found that the blockade of glycolysis in mouse brain with 2-deoxyglucose administration resulted in decreased transcellular permeability of the BBB. In conclusion, utilizing glycolysis in brain ECs has critical roles in the maintenance and permeability of the BBB. Overall, we could conclude that brain ECs are highly glycolytic, and their energy can be used to maintain the transcellular permeability of the BBB.
Collapse
|
23
|
Mechanisms of selective monocyte targeting by liposomes functionalized with a cationic, arginine-rich lipopeptide. Acta Biomater 2022; 144:96-108. [PMID: 35314364 DOI: 10.1016/j.actbio.2022.03.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 01/01/2023]
Abstract
Stimulation of monocytes with immunomodulating agents can harness the immune system to treat a long range of diseases, including cancers, infections and autoimmune diseases. To this end we aimed to develop a monocyte-targeting delivery platform based on cationic liposomes, which can be utilized to deliver immunomodulators and thus induce monocyte-mediated immune responses while avoiding off-target side-effects. The cationic liposome design is based on functionalizing the liposomal membrane with a cholesterol-anchored tri-arginine peptide (TriArg). We demonstrate that TriArg liposomes can target monocytes with high specificity in both human and murine blood and that this targeting is dependent on the content of TriArg in the liposomal membrane. In addition, we show that the mechanism of selective monocyte targeting involves the CD14 co-receptor, and selectivity is compromised when the TriArg content is increased, resulting in complement-mediated off-target uptake in granulocytes. The presented mechanistic findings of uptake by peripheral blood leukocytes may guide the design of future drug delivery systems utilized for immunotherapy. STATEMENT OF SIGNIFICANCE: Monocytes are attractive targets for immunotherapies of cancers, infections and autoimmune diseases. Specific delivery of immunostimulatory drugs to monocytes is typically achieved using ligand-targeted drug delivery systems, but a simpler approach is to target monocytes using cationic liposomes. To achieve this, however, a deep understanding of the mechanisms governing the interactions of cationic liposomes with monocytes and other leukocytes is required. We here investigate these interactions using liposomes incorporating a cationic arginine-rich lipopeptide. We demonstrate that monocyte targeting can be achieved by fine-tuning the lipopeptide content in the liposomes. Additionally, we reveal that the CD14 receptor is involved in the targeting process, whereas the complement system is not. These mechanistic findings are critical for future design of monocyte-targeting liposomal therapies.
Collapse
|
24
|
Brancolini G, Rotello VM, Corni S. Role of Ionic Strength in the Formation of Stable Supramolecular Nanoparticle-Protein Conjugates for Biosensing. Int J Mol Sci 2022; 23:ijms23042368. [PMID: 35216496 PMCID: PMC8874478 DOI: 10.3390/ijms23042368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022] Open
Abstract
Monolayer-protected gold nanoparticles (AuNPs) exhibit distinct physical and chemical properties depending on the nature of the ligand chemistry. A commonly employed NP monolayer comprises hydrophobic molecules linked to a shell of PEG and terminated with functional end group, which can be charged or neutral. Different layers of the ligand shell can also interact in different manners with proteins, expanding the range of possible applications of these inorganic nanoparticles. AuNP-fluorescent Green Fluorescent Protein (GFP) conjugates are gaining increasing attention in sensing applications. Experimentally, their stability is observed to be maintained at low ionic strength conditions, but not at physiologically relevant conditions of higher ionic strength, limiting their applications in the field of biosensors. While a significant amount of fundamental work has been done to quantify electrostatic interactions of colloidal nanoparticle at the nanoscale, a theoretical description of the ion distribution around AuNPs still remains relatively unexplored. We perform extensive atomistic simulations of two oppositely charged monolayer-protected AuNPs interacting with fluorescent supercharged GFPs co-engineered to have complementary charges. These simulations were run at different ionic strengths to disclose the role of the ionic environment on AuNP–GFP binding. The results highlight the capability of both AuNPs to intercalate ions and water molecules within the gold–sulfur inner shell and the different tendency of ligands to bend inward allowing the protein to bind not only with the terminal ligands but also the hydrophobic alkyl chains. Different binding stability is observed in the two investigated cases as a function of the ligand chemistry.
Collapse
Affiliation(s)
- Giorgia Brancolini
- Institute of Nanoscience, CNR-NANO S3, via G. Campi 213/A, 41125 Modena, Italy;
- Correspondence: ; Tel.: +39-059-2055333
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA;
| | - Stefano Corni
- Institute of Nanoscience, CNR-NANO S3, via G. Campi 213/A, 41125 Modena, Italy;
- Department of Chemical Sciences, University of Padova, via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
25
|
Ray M, Brancolini G, Luther DC, Jiang Z, Cao-Milán R, Cuadros AM, Burden A, Clark V, Rana S, Mout R, Landis RF, Corni S, Rotello VM. High affinity protein surface binding through co-engineering of nanoparticles and proteins. NANOSCALE 2022; 14:2411-2418. [PMID: 35089292 PMCID: PMC8941649 DOI: 10.1039/d1nr07497k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Control over supramolecular recognition between proteins and nanoparticles (NPs) is of fundamental importance in therapeutic applications and sensor development. Most NP-protein binding approaches use 'tags' such as biotin or His-tags to provide high affinity; protein surface recognition provides a versatile alternative strategy. Generating high affinity NP-protein interactions is challenging however, due to dielectric screening at physiological ionic strengths. We report here the co-engineering of nanoparticles and protein to provide high affinity binding. In this strategy, 'supercharged' proteins provide enhanced interfacial electrostatic interactions with complementarily charged nanoparticles, generating high affinity complexes. Significantly, the co-engineered protein-nanoparticle assemblies feature high binding affinity even at physiologically relevant ionic strength conditions. Computational studies identify both hydrophobic and electrostatic interactions as drivers for these high affinity NP-protein complexes.
Collapse
Affiliation(s)
- Moumita Ray
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Giorgia Brancolini
- Center S3, CNR Institute of Nanoscience, via Campi 213/A, 41125 Modena, Italy
| | - David C Luther
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Ziwen Jiang
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Roberto Cao-Milán
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Alejandro M Cuadros
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Andrew Burden
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Vincent Clark
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Subinoy Rana
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Rubul Mout
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Ryan F Landis
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| | - Stefano Corni
- Center S3, CNR Institute of Nanoscience, via Campi 213/A, 41125 Modena, Italy
- Department of Chemical Science, University of Padova, Via Francesco Marzolo 1, 35131 Padova, Italy
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA.
| |
Collapse
|
26
|
Ebrahimi SB, Samanta D, Kusmierz CD, Mirkin CA. Protein transfection via spherical nucleic acids. Nat Protoc 2022; 17:327-357. [PMID: 35039669 DOI: 10.1038/s41596-021-00642-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023]
Abstract
The efficient transfection of functional proteins into cells can serve as a means for regulating cellular processes toward solving fundamental challenges in biology and medicine. However, the use of proteins as effective intracellular agents is hindered by their low cellular uptake and susceptibility to degradation. Over the past 15 years, our group has been developing spherical nucleic acids (SNAs), nanoparticles functionalized with a dense radially oriented shell of nucleic acids. These structures actively enter cells and have opened new frontiers in chemical sensing, biodiagnostics and therapeutics. Recently, we have shown that proteins can be used as structurally precise and homogeneous nanoparticle cores in SNAs. The resultant protein SNAs (ProSNAs) allow previously cell-impermeable proteins to actively enter cells, exhibit high degrees of stability and activity both in cell culture and in vivo, and show enhanced pharmacokinetics. Consequently, these modular structures constitute a plug-and-play platform in which the protein core and nucleic acid shell can be independently varied to achieve a desired function. Here, we describe the synthesis of ProSNAs through the chemical modification of solvent-accessible surface residues (3-5 d). We also discuss design considerations, strategies for characterization, and applications of ProSNAs in cellular transfection, biological sensing and functional enzyme delivery in vivo.
Collapse
Affiliation(s)
- Sasha B Ebrahimi
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA
| | - Devleena Samanta
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Caroline D Kusmierz
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Chad A Mirkin
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA.
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
27
|
Chen M, Wang H, Guo H, Zhang Y, Chen L. Systematic Investigation of Biocompatible Cationic Polymeric Nucleic Acid Carriers for Immunotherapy of Hepatocellular Carcinoma. Cancers (Basel) 2021; 14:85. [PMID: 35008249 PMCID: PMC8750096 DOI: 10.3390/cancers14010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third-largest cause of cancer death worldwide, while immunotherapy is rapidly being developed to fight HCC with great potential. Nucleic acid drugs are the most important modulators in HCC immunotherapy. To boost the efficacy of therapeutics and amplify the efficiency of genetic materials, biocompatible polymers are commonly used. However, under the strong need of a summary for current developments of biocompatible polymeric nucleic acid carriers for immunotherapy of HCC, there is rare review article specific to this topic to our best knowledge. In this article, we will discuss the current progress of immunotherapy for HCC, biocompatible cationic polymers (BCPs) as nucleic acid carriers used (or potential) to fight HCC, the roles of biocompatible polymeric carriers for nucleic acid delivery, and nucleic acid delivery by biocompatible polymers for immunotherapy. At the end, we will conclude the review and discuss future perspectives. This article discusses biocompatible polymeric nucleic acid carriers for immunotherapy of HCC from multidiscipline perspectives and provides a new insight in this domain. We believe this review will be interesting to polymer chemists, pharmacists, clinic doctors, and PhD students in related disciplines.
Collapse
Affiliation(s)
- Mingsheng Chen
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| | - Hao Wang
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| | - Hongying Guo
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| | - Ying Zhang
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Liang Chen
- Shanghai Public Health Clinic Center, Fudan University, Shanghai 201508, China; (M.C.); (H.W.); (H.G.)
| |
Collapse
|
28
|
Ma K, Li X, Xu B, Tian W. Label-free bioassay with graphene oxide-based fluorescent aptasensors: A review. Anal Chim Acta 2021; 1188:338859. [PMID: 34794573 DOI: 10.1016/j.aca.2021.338859] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
Bioassays using a fluorophore and DNA aptamer have been extensively developed due to the ultrasensitivity of fluorophores and recognition ability of DNA aptamers. Conventional fluorescent aptamer-based sensors (aptasensors) require chemical labeling between the fluorophore and aptamer and is technologically impracical for various sensing and assay applications. A simple "mix and go" strategy has been introduced that uses label-free technology as a platform for sensor development. The biosensors comprise a fluorophore, a ssDNA aptamer, and eco-friendly graphene oxide (GO). In the absence of the sensor target, GO quenches the fluorescence of the fluorophore and single-strand DNA aptamer complex. When the target is added, the DNA aptamer conformationally turns into a duplex, G-quadruplexe, or other secondary structure. This structure change leads to release of GO by the fluorophore-aptamer-target complex, generating dramatic fluorescence recovery and amplification. With this sensing method, the DNA aptamer does not need to be chemically labeled. Therefore, flexible fluorophore indicators and ssDNA aptamers can be used in this label-free aptasensing strategy. In this review, we discuss various unlabeled fluorophores, including synthetic small molecular fluorophores and genetically encoded fluorescent proteins, as indicators for generating GO-based fluorescent DNA aptasensors for label-free bioassay.
Collapse
Affiliation(s)
- Ke Ma
- Center of AIE Research, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, PR China
| | - Xing Li
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, PR China.
| | - Bin Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Wenjing Tian
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| |
Collapse
|
29
|
Horn JM, Obermeyer AC. Genetic and Covalent Protein Modification Strategies to Facilitate Intracellular Delivery. Biomacromolecules 2021; 22:4883-4904. [PMID: 34855385 PMCID: PMC9310055 DOI: 10.1021/acs.biomac.1c00745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein-based therapeutics represent a rapidly growing segment of approved disease treatments. Successful intracellular delivery of proteins is an important precondition for expanded in vivo and in vitro applications of protein therapeutics. Direct modification of proteins and peptides for improved cytosolic translocation are a promising method of increasing delivery efficiency and expanding the viability of intracellular protein therapeutics. In this Review, we present recent advances in both synthetic and genetic protein modifications for intracellular delivery. Active endocytosis-based and passive internalization pathways are discussed, followed by a review of modification methods for improved cytosolic delivery. After establishing how proteins can be modified, general strategies for facilitating intracellular delivery, such as chemical supercharging or inclusion of cell-penetrating motifs, are covered. We then outline protein modifications that promote endosomal escape. We finally examine the delivery of two potential classes of therapeutic proteins, antibodies and associated antibody fragments, and gene editing proteins, such as cas9.
Collapse
|
30
|
Bulos JA, Guo R, Wang Z, DeLessio MA, Saven JG, Dmochowski IJ. Design of a Superpositively Charged Enzyme: Human Carbonic Anhydrase II Variant with Ferritin Encapsulation and Immobilization. Biochemistry 2021; 60:3596-3609. [PMID: 34757723 DOI: 10.1021/acs.biochem.1c00515] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Supercharged proteins exhibit high solubility and other desirable properties, but no engineered superpositively charged enzymes have previously been made. Superpositively charged variants of proteins such as green fluorescent protein have been efficiently encapsulated within Archaeoglobus fulgidus thermophilic ferritin (AfFtn). Encapsulation by supramolecular ferritin can yield systems with a variety of sequestered cargo. To advance applications in enzymology and green chemistry, we sought a general method for supercharging an enzyme that retains activity and is compatible with AfFtn encapsulation. The zinc metalloenzyme human carbonic anhydrase II (hCAII) is an attractive encapsulation target based on its hydrolytic activity and physiologic conversion of carbon dioxide to bicarbonate. A computationally designed variant of hCAII contains positively charged residues substituted at 19 sites on the protein's surface, resulting in a shift of the putative net charge from -1 to +21. This designed hCAII(+21) exhibits encapsulation within AfFtn without the need for fusion partners or additional reagents. The hCAII(+21) variant retains esterase activity comparable to the wild type and spontaneously templates the assembly of AfFtn 24mers around itself. The AfFtn-hCAII(+21) host-guest complex exhibits both greater activity and thermal stability when compared to hCAII(+21). Upon immobilization on a solid support, AfFtn-hCAII(+21) retains enzymatic activity and exhibits an enhancement of activity at elevated temperatures.
Collapse
Affiliation(s)
- Joshua A Bulos
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Rui Guo
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Zhiheng Wang
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Maegan A DeLessio
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jeffery G Saven
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ivan J Dmochowski
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
31
|
Martinent R, Tawffik S, López-Andarias J, Moreau D, Laurent Q, Matile S. Dithiolane quartets: thiol-mediated uptake enables cytosolic delivery in deep tissue. Chem Sci 2021; 12:13922-13929. [PMID: 34760179 PMCID: PMC8549803 DOI: 10.1039/d1sc04828g] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
The cytosolic delivery of various substrates in 3D multicellular spheroids by thiol-mediated uptake is reported. This is important because most orthodox systems, including polycationic cell-penetrating peptides, fail to deliver efficiently into deep tissue. The grand principles of supramolecular chemistry, that is the pH dependence of dynamic covalent disulfide exchange with known thiols on the transferrin receptor, are proposed to account for transcytosis into deep tissue, while the known but elusive exchange cascades along the same or other partners assure cytosolic delivery in kinetic competition. For quantitative detection in the cytosol, the 2D chloroalkane penetration assay (CAPA) is translated to 3D deep tissue. The targeted delivery of quantum dots, otherwise already troublesome in 2D culture, and the controlled release of mechanophores are realized to exemplify the power of thiol-mediated uptake into spheroids. As transporters, dithiolane quartets on streptavidin templates are introduced as modular motifs. Built from two amino acids only, the varied stereochemistry and peptide sequence are shown to cover maximal functional space with minimal structural change, i.e., constitutional isomers. Reviving a classic in peptide chemistry, this templated assembly of β quartets promises to expand streptavidin biotechnology in new directions, while the discovery of general cytosolic delivery in deep tissue as an intrinsic advantage further enhances the significance and usefulness of thiol-mediated uptake.
Collapse
Affiliation(s)
- Rémi Martinent
- Department of Organic Chemistry, University of Geneva Geneva Switzerland https://www.unige.ch/sciences/chiorg/matile/ +41 22 379 6523
| | - Salman Tawffik
- Department of Organic Chemistry, University of Geneva Geneva Switzerland https://www.unige.ch/sciences/chiorg/matile/ +41 22 379 6523
| | - Javier López-Andarias
- Department of Organic Chemistry, University of Geneva Geneva Switzerland https://www.unige.ch/sciences/chiorg/matile/ +41 22 379 6523
| | - Dimitri Moreau
- Department of Organic Chemistry, University of Geneva Geneva Switzerland https://www.unige.ch/sciences/chiorg/matile/ +41 22 379 6523
| | - Quentin Laurent
- Department of Organic Chemistry, University of Geneva Geneva Switzerland https://www.unige.ch/sciences/chiorg/matile/ +41 22 379 6523
| | - Stefan Matile
- Department of Organic Chemistry, University of Geneva Geneva Switzerland https://www.unige.ch/sciences/chiorg/matile/ +41 22 379 6523
| |
Collapse
|
32
|
Sun H, Zhu X, Li C, Ma Z, Han X, Luo Y, Yang L, Yu J, Miao Y. Xanthomonas effector XopR hijacks host actin cytoskeleton via complex coacervation. Nat Commun 2021; 12:4064. [PMID: 34210966 PMCID: PMC8249405 DOI: 10.1038/s41467-021-24375-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 06/11/2021] [Indexed: 01/09/2023] Open
Abstract
The intrinsically disordered region (IDR) is a preserved signature of phytobacterial type III effectors (T3Es). The T3E IDR is thought to mediate unfolding during translocation into the host cell and to avoid host defense by sequence diversification. Here, we demonstrate a mechanism of host subversion via the T3E IDR. We report that the Xanthomonas campestris T3E XopR undergoes liquid-liquid phase separation (LLPS) via multivalent IDR-mediated interactions that hijack the Arabidopsis actin cytoskeleton. XopR is gradually translocated into host cells during infection and forms a macromolecular complex with actin-binding proteins at the cell cortex. By tuning the physical-chemical properties of XopR-complex coacervates, XopR progressively manipulates multiple steps of actin assembly, including formin-mediated nucleation, crosslinking of F-actin, and actin depolymerization, which occurs through competition for actin-depolymerizing factor and depends on constituent stoichiometry. Our findings unravel a sophisticated strategy in which bacterial T3E subverts the host actin cytoskeleton via protein complex coacervation.
Collapse
Affiliation(s)
- He Sun
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xinlu Zhu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Chuanxi Li
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Zhiming Ma
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Xiao Han
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yuanyuan Luo
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Liang Yang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jing Yu
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Yansong Miao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
33
|
An artificial membrane binding protein-polymer surfactant nanocomplex facilitates stem cell adhesion to the cartilage extracellular matrix. Biomaterials 2021; 276:120996. [PMID: 34280823 DOI: 10.1016/j.biomaterials.2021.120996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/12/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
One of the major challenges within the emerging field of injectable stem cell therapies for articular cartilage (AC) repair is the retention of sufficient viable cell numbers at the site of injury. Even when delivered via intra-articular injection, the number of stem cells retained at the target is often low and declines rapidly over time. To address this challenge, an artificial plasma membrane binding nanocomplex was rationally designed to provide human mesenchymal stem cells (hMSCs) with increased adhesion to articular cartilage tissue. The nanocomplex comprises the extracellular matrix (ECM) binding peptide of a placenta growth factor-2 (PlGF-2) fused to a supercharged green fluorescent protein (scGFP), which was electrostatically conjugated to anionic polymer surfactant chains to yield [S-]scGFP_PlGF2. The [S-]scGFP_PlGF2 nanocomplex spontaneously inserts into the plasma membrane of hMSCs, is not cytotoxic, and does not inhibit differentiation. The nanocomplex-modified hMSCs showed a significant increase in affinity for immobilised collagen II, a key ECM protein of cartilage, in both static and dynamic cell adhesion assays. Moreover, the cells adhered strongly to bovine ex vivo articular cartilage explants resulting in high cell numbers. These findings suggest that the re-engineering of hMSC membranes with [S-]scGFP_PlGF2 could improve the efficacy of injectable stem cell-based therapies for the treatment of damaged articular cartilage.
Collapse
|
34
|
Hango CR, Backlund CM, Davis HC, Posey ND, Minter LM, Tew GN. Non-Covalent Carrier Hydrophobicity as a Universal Predictor of Intracellular Protein Activity. Biomacromolecules 2021; 22:2850-2863. [PMID: 34156837 DOI: 10.1021/acs.biomac.1c00242] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Over the past decade, extensive optimization of polymeric cell-penetrating peptide (CPP) mimics (CPPMs) by our group has generated a substantial library of broadly effective carriers which circumvent the need for covalent conjugation often required by CPPs. In this study, design rules learned from CPPM development were applied to reverse-engineer the first library of simple amphiphilic block copolypeptides for non-covalent protein delivery, namely, poly(alanine-block-arginine), poly(phenylalanine-block-arginine), and poly(tryptophan-block-arginine). This new CPP library was screened for enhanced green fluorescent protein and Cre recombinase delivery alongside a library of CPPMs featuring equivalent side-chain configurations. Due to the added hydrophobicity imparted by the polymer backbone as compared to the polypeptide backbone, side-chain functionality was not a universal predictor of carrier performance. Rather, overall carrier hydrophobicity predicted the top performers for both internalization and activity of protein cargoes, regardless of backbone identity. Furthermore, comparison of protein uptake and function revealed carriers which facilitated high gene recombination despite remarkably low Cre internalization, leading us to formalize the concept of intracellular availability (IA) of the delivered cargo. IA, a measure of cargo activity per quantity of cargo internalized, provides valuable insight into the physical relationship between cellular internalization and bioavailability, which can be affected by bottlenecks such as endosomal escape and cargo release. Importantly, carriers with maximal IA existed within a narrow hydrophobicity window, more hydrophilic than those exhibiting maximal cargo uptake. Hydrophobicity may be used as a scaffold-independent predictor of protein uptake, function, and IA, enabling identification of new, effective carriers which would be overlooked by uptake-based screening methods.
Collapse
Affiliation(s)
- Christopher R Hango
- Department of Polymer Science & Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Coralie M Backlund
- Department of Polymer Science & Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Hazel C Davis
- Department of Polymer Science & Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Nicholas D Posey
- Department of Polymer Science & Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Lisa M Minter
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States.,Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, Untied States
| | - Gregory N Tew
- Department of Polymer Science & Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, United States.,Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, Untied States
| |
Collapse
|
35
|
Kasahara K, Kuroda D, Tanabe A, Kawade R, Nagatoishi S, Tsumoto K. Anion solvation enhanced by positive supercharging mutations preserves thermal stability of an antibody in a wide pH range. Biochem Biophys Res Commun 2021; 563:54-59. [PMID: 34058475 DOI: 10.1016/j.bbrc.2021.05.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 05/16/2021] [Indexed: 12/27/2022]
Abstract
Proteins function through interactions with other molecules. In protein engineering, scientists often engineer proteins by mutating their amino acid sequences on the protein surface to improve various physicochemical properties. "Supercharging" is a method to design proteins by mutating surface residues with charged amino acids. Previous studies demonstrated that supercharging mutations conferred better thermal resistance, solubility, and cell penetration to proteins. Likewise, antibodies recognize antigens through the antigen-binding site on the surface. The genetic and structural diversity of antibodies leads to high specificity and affinity toward antigens, enabling antibodies to be versatile tools in various applications. When assessing therapeutic antibodies, surface charge is an important factor to consider because the isoelectric point plays a role in protein clearance inside the body. In this study, we explored how supercharging mutations affect physicochemical properties of antibodies. Starting from a crystal structure of an antibody with the net charge of -4, we computationally designed a supercharged variant possessing the net charge of +10. The positive-supercharged antibody exhibited marginal improvement in thermal stability, but the secondary structure and the binding affinity to the antigen (net charge of +8) were preserved. We also used physicochemical measurements and molecular dynamics simulations to analyze the effects of supercharging mutations in sodium phosphate buffer with different pH and ion concentrations, which revealed preferential solvation of phosphate ions to the supercharged surface relative to the wild-type surface. These results suggest that supercharging would be a useful approach to preserving thermal stability of antibodies in a wide range of pH, which may enable further diversification of antibody repertoires beyond natural evolution.
Collapse
Affiliation(s)
- Keisuke Kasahara
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Daisuke Kuroda
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| | - Aki Tanabe
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Raiji Kawade
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
36
|
Levasseur MD, Mantri S, Hayashi T, Reichenbach M, Hehn S, Waeckerle-Men Y, Johansen P, Hilvert D. Cell-Specific Delivery Using an Engineered Protein Nanocage. ACS Chem Biol 2021; 16:838-843. [PMID: 33881303 DOI: 10.1021/acschembio.1c00007] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanoparticle-based delivery systems have shown great promise for theranostics and bioimaging on the laboratory scale due to favorable pharmacokinetics and biodistribution. In this study, we examine the utility of a cage-forming variant of the protein lumazine synthase, which was previously designed and evolved to encapsulate biomacromolecular cargo. Linking antibody-binding domains to the exterior of the cage enabled binding of targeting immunoglobulins and cell-specific uptake of encapsulated cargo. Protein nanocages displaying antibody-binding domains appear to be less immunogenic than their unmodified counterparts, but they also recruit serum antibodies that can mask the efficacy of the targeting antibody. Our study highlights the strengths and limitations of a common targeting strategy for practical nanoparticle-based delivery applications.
Collapse
Affiliation(s)
| | - Shiksha Mantri
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Takahiro Hayashi
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Maria Reichenbach
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Svenja Hehn
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | | | - Pål Johansen
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
37
|
Knox S, Wissner R, Piszkiewicz S, Schepartz A. Cytosolic Delivery of Argininosuccinate Synthetase Using a Cell-Permeant Miniature Protein. ACS CENTRAL SCIENCE 2021; 7:641-649. [PMID: 34056094 PMCID: PMC8155463 DOI: 10.1021/acscentsci.0c01603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Indexed: 05/04/2023]
Abstract
Citrullinemia type I (CTLN-I) results from the absence or deficiency of argininosuccinate synthetase (AS), a 46 kDa enzyme that acts in the cytosol of hepatocytes to convert aspartic acid and citrulline into argininosuccinic acid. AS is an essential component of the urea cycle, and its absence or deficiency results in the harmful accumulation of ammonia in blood and cerebrospinal fluid. No disease-modifying treatment of CTLN-I exists. Here we report that the cell-permeant miniature protein (CPMP) ZF5.3 (ZF) can deliver AS to the cytosol of cells in culture and the livers of healthy mice. The fusion protein ZF-AS is catalytically active in vitro, stabilized in plasma, and traffics successfully to the cytosol of cultured Saos-2 and SK-HEP-1 cells, achieving cytosolic concentrations greater than 100 nM. This value is 3-10-fold higher than the concentration of endogenous AS (11 ± 1 to 44 ± 5 nM). When injected into healthy C57BL/6 mice, ZF-AS reaches the mouse liver to establish concentrations almost 200 nM above baseline. These studies demonstrate that ZF5.3 can deliver a complex enzyme to the cytosol at therapeutically relevant concentrations and support its application as an improved delivery vehicle for therapeutic proteins that function in the cytosol, including enzyme replacement therapies.
Collapse
Affiliation(s)
- Susan
L. Knox
- Department
of Chemistry, Yale University, New Haven, Connecticut 06511, United States
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
| | - Rebecca Wissner
- Department
of Chemistry, Yale University, New Haven, Connecticut 06511, United States
| | - Samantha Piszkiewicz
- Department
of Chemistry, Yale University, New Haven, Connecticut 06511, United States
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
| | - Alanna Schepartz
- Department
of Chemistry, University of California, Berkeley, California 94720, United States
- Department
of Molecular and Cell Biology, University
of California, Berkeley, California 94720, United States
- California
Institute for Quantitative Biosciences, University of California, Berkeley, California 94720, United States
- E-mail:
| |
Collapse
|
38
|
Yang Z, Lee MMM, Chan MK. Efficient intracellular delivery of p53 protein by engineered protein crystals restores tumor suppressing function in vivo. Biomaterials 2021; 271:120759. [PMID: 33798968 DOI: 10.1016/j.biomaterials.2021.120759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
Direct delivery of proteins into cells holds significant potential for basic research and drug development. However, the poor endosomal escape of conventional delivery strategies remains a challenge, thus limiting the clinical translation of many protein therapeutics. Herein, we report that engineered Cry3Aa protein (Pos3Aa) crystals formed naturally within Bacillus thuringiensis can serve as a vehicle for efficient cytosolic delivery of bioactive proteins. We showed that Pos3Aa-mediated delivery of tumor suppressor p53 protein, a promising therapeutic candidate found to be inactivated in nearly half of human cancers, resulted in the restoration of p53 function in p53-deficient cancer cells, and thereby sensitized them to 5-fluorouracil chemotherapy as demonstrated in in vitro and in vivo models. Our results validate that Pos3Aa crystals can be a robust and effective platform for the cytosolic delivery of effector proteins, and suggest that efficient uptake and endosomal escape could be critical for efficacious p53 protein-based cancer therapy.
Collapse
Affiliation(s)
- Zaofeng Yang
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Marianne M M Lee
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Michael K Chan
- School of Life Sciences and Center of Novel Biomaterials, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
39
|
Garcia J, Fernández‐Pradas JM, Lladó A, Serra P, Zalvidea D, Kogan MJ, Giralt E, Sánchez‐Navarro M. The Combined Use of Gold Nanoparticles and Infrared Radiation Enables Cytosolic Protein Delivery. Chemistry 2021; 27:4670-4675. [DOI: 10.1002/chem.202005000] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Josep Garcia
- Institute for Research in Biomedicine Barcelona Institute of, Science and Technology Baldiri Reixac 10 08028 Barcelona Spain
| | - J. Marcos Fernández‐Pradas
- Department of Applied Physics University of Barcelona Martí i Franquès 1 08028 Barcelona Spain
- Institute of Nanoscience and Nanotechnology (IN2UB) University of Barcelona Av. Diagonal 645 08028 Barcelona Spain
| | - Anna Lladó
- Institute for Research in Biomedicine Barcelona Institute of, Science and Technology Baldiri Reixac 10 08028 Barcelona Spain
| | - Pere Serra
- Department of Applied Physics University of Barcelona Martí i Franquès 1 08028 Barcelona Spain
- Institute of Nanoscience and Nanotechnology (IN2UB) University of Barcelona Av. Diagonal 645 08028 Barcelona Spain
| | - Dobryna Zalvidea
- Institute for Bioengineering of Catalonia (IBEC) Barcelona Institute of Technology (BIST) Barcelona Spain
| | - Marcelo J. Kogan
- Departamento de Química Farmacológica y Toxicológica Facultad de Ciencias Químicas y Farmacéuticas Universidad de Chile Santiago Chile
- Advanced Center for Chronic Diseases (ACCDiS) Sergio Livingstone 1007, Independencia Santiago Chile
| | - Ernest Giralt
- Institute for Research in Biomedicine Barcelona Institute of, Science and Technology Baldiri Reixac 10 08028 Barcelona Spain
- Department of Inorganic and Organic Chemistry University of Barcelona Martí i Franquès 1–11 08028 Barcelona Spain
| | - Macarena Sánchez‐Navarro
- Institute for Research in Biomedicine Barcelona Institute of, Science and Technology Baldiri Reixac 10 08028 Barcelona Spain
| |
Collapse
|
40
|
Anson F, Kanjilal P, Thayumanavan S, Hardy JA. Tracking exogenous intracellular casp-3 using split GFP. Protein Sci 2021; 30:366-380. [PMID: 33165988 PMCID: PMC7784757 DOI: 10.1002/pro.3992] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/28/2020] [Accepted: 11/03/2020] [Indexed: 11/08/2022]
Abstract
Cytosolic protein delivery promises diverse applications from therapeutics, to genetic modification and precision research tools. To achieve effective cellular and subcellular delivery, approaches that allow protein visualization and accurate localization with greater sensitivity are essential. Fluorescently tagging proteins allows detection, tracking and visualization in cellulo. However, undesired consequences from fluorophores or fluorescent protein tags, such as nonspecific interactions and high background or perturbation to native protein's size and structure, are frequently observed, or more troublingly, overlooked. Distinguishing cytosolically released molecules from those that are endosomally entrapped upon cellular uptake is particularly challenging and is often complicated by the inherent pH-sensitive and hydrophobic properties of the fluorophore. Monitoring localization is more complex in delivery of proteins with inherent protein-modifying activities like proteases, transacetylases, kinases, etc. Proteases are among the toughest cargos due to their inherent propensity for self-proteolysis. To implement a reliable, but functionally silent, tagging technology in a protease, we have developed a caspase-3 variant tagged with the 11th strand of GFP that retains both enzymatic activity and structural characteristics of wild-type caspase-3. Only in the presence of cytosolic GFP strands 1-10 will the tagged caspase-3 generate fluorescence to signal a non-endosomal location. This methodology facilitates easy screening of cytosolic vs. endosomally-entrapped proteins due to low probabilities for false positive results, and further, allows tracking of the resultant cargo's translocation. The development of this tagged casp-3 cytosolic reporter lays the foundation to probe caspase therapeutic properties, charge-property relationships governing successful escape, and the precise number of caspases required for apoptotic cell death.
Collapse
Affiliation(s)
- Francesca Anson
- Department of ChemistryUniversity of MassachusettsAmherstMassachusettsUSA
| | - Pintu Kanjilal
- Department of ChemistryUniversity of MassachusettsAmherstMassachusettsUSA
| | - S. Thayumanavan
- Department of ChemistryUniversity of MassachusettsAmherstMassachusettsUSA
- The Center for Bioactive Delivery at the Institute for Applied Life SciencesUniversity of MassachusettsAmherstMassachusettsUSA
| | - Jeanne A. Hardy
- Department of ChemistryUniversity of MassachusettsAmherstMassachusettsUSA
- The Center for Bioactive Delivery at the Institute for Applied Life SciencesUniversity of MassachusettsAmherstMassachusettsUSA
| |
Collapse
|
41
|
Patino CA, Mukherjee P, Lemaitre V, Pathak N, Espinosa HD. Deep Learning and Computer Vision Strategies for Automated Gene Editing with a Single-Cell Electroporation Platform. SLAS Technol 2021; 26:26-36. [PMID: 33449846 DOI: 10.1177/2472630320982320] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Single-cell delivery platforms like microinjection and nanoprobe electroporation enable unparalleled control over cell manipulation tasks but are generally limited in throughput. Here, we present an automated single-cell electroporation system capable of automatically detecting cells with artificial intelligence (AI) software and delivering exogenous cargoes of different sizes with uniform dosage. We implemented a fully convolutional network (FCN) architecture to precisely locate the nuclei and cytosol of six cell types with various shapes and sizes, using phase contrast microscopy. Nuclear staining or reporter fluorescence was used along with phase contrast images of cells within the same field of view to facilitate the manual annotation process. Furthermore, we leveraged the near-human inference capabilities of the FCN network in detecting stained nuclei to automatically generate ground-truth labels of thousands of cells within seconds, and observed no statistically significant difference in performance compared to training with manual annotations. The average detection sensitivity and precision of the FCN network were 95±1.7% and 90±1.8%, respectively, outperforming a traditional image-processing algorithm (72±7.2% and 72±5.5%) used for comparison. To test the platform, we delivered fluorescent-labeled proteins into adhered cells and measured a delivery efficiency of 90%. As a demonstration, we used the automated single-cell electroporation platform to deliver Cas9-guide RNA (gRNA) complexes into an induced pluripotent stem cell (iPSC) line to knock out a green fluorescent protein-encoding gene in a population of ~200 cells. The results demonstrate that automated single-cell delivery is a useful cell manipulation tool for applications that demand throughput, control, and precision.
Collapse
Affiliation(s)
- Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.,iNfinitesimal LLC, Skokie, IL, USA
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.,Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, USA.,iNfinitesimal LLC, Skokie, IL, USA
| | | | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.,Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, USA
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.,Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, USA.,iNfinitesimal LLC, Skokie, IL, USA
| |
Collapse
|
42
|
Congdon MD, Gildersleeve JC. Enhanced Binding and Reduced Immunogenicity of Glycoconjugates Prepared via Solid-State Photoactivation of Aliphatic Diazirine Carbohydrates. Bioconjug Chem 2020; 32:133-142. [PMID: 33325683 DOI: 10.1021/acs.bioconjchem.0c00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Biological conjugation is an important tool employed for many basic research and clinical applications. While useful, common methods of biological conjugation suffer from a variety of limitations, such as (a) requiring the presence of specific surface-exposed residues, such as lysines or cysteines, (b) reducing protein activity, and/or (c) reducing protein stability and solubility. Use of photoreactive moieties including diazirines, azides, and benzophenones provide an alternative, mild approach to conjugation. Upon irradiation with UV and visible light, these functionalities generate highly reactive carbenes, nitrenes, and radical intermediates. Many of these will couple to proteins in a non-amino-acid-specific manner. The main hurdle for photoactivated biological conjugation is very low yield. In this study, we developed a solid-state method to increase conjugation efficiency of diazirine-containing carbohydrates to proteins. Using this methodology, we produced multivalent carbohydrate-protein conjugates with unaltered protein charge and secondary structure. Compared to carbohydrate conjugates prepared with amide linkages to lysine residues using standard NHS conjugation, the photoreactive prepared conjugates displayed up to 100-fold improved binding to lectins and diminished immunogenicity in mice. These results indicate that photoreactive bioconjugation could be especially useful for in vivo applications, such as lectin targeting, where high binding affinity and low immunogenicity are desired.
Collapse
Affiliation(s)
- Molly D Congdon
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Jeffrey C Gildersleeve
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| |
Collapse
|
43
|
Abstract
Diffusivity of a protein (a Brownian particle) is caused by random molecular collisions in the Stokes-Einstein picture. Alternatively, it can be viewed as driven by unbalanced stochastic forces acting from water on the protein. Molecular dynamics simulations of protein mutants carrying different charges are analyzed here in terms of the van der Waals (vdW) and electrostatic forces acting on the protein. They turn out to be remarkably strongly correlated and the total force is largely a compensation between vdW and electrostatic forces. Both vdW and electrostatic forces relax on the same time scale of 5-6 ns separated by 6 orders of magnitude from the relaxation time of the total force. Similar phenomenology applies to the dynamics and statistics of the fluctuating torque responsible for rotational diffusion. Standard linear theories of dielectric friction are grossly inapplicable to translational and rotational diffusion of proteins overestimating friction by many orders of magnitude.
Collapse
Affiliation(s)
- Setare Mostajabi Sarhangi
- Department of Physics and School of Molecular Sciences, Arizona State University, P.O. Box 871504, Tempe, Arizona 85287-1504, United States
| | - Dmitry V Matyushov
- Department of Physics and School of Molecular Sciences, Arizona State University, P.O. Box 871504, Tempe, Arizona 85287-1504, United States
| |
Collapse
|
44
|
Takikawa M, Fujisawa M, Yoshino K, Takeoka S. Intracellular Distribution of Lipids and Encapsulated Model Drugs from Cationic Liposomes with Different Uptake Pathways. Int J Nanomedicine 2020; 15:8401-8409. [PMID: 33149583 PMCID: PMC7605631 DOI: 10.2147/ijn.s267638] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/31/2020] [Indexed: 01/06/2023] Open
Abstract
AIM The uptake pathway of liposomes into cells is mainly via endocytosis or membrane fusion; however, the relationship between the uptake pathway and the intracellular pharmacokinetics of the liposome components remains unclear. This study aimed at revealing the relationship by using cationic liposomes having similar physical properties and different uptake pathways. MATERIALS AND METHODS We prepared cationic liposomes composed of amino acid-type lipids, K3C14 and K3C16, which have different uptake pathways by a hydration method, and fluorescently modified them by encapsulating FITC-dextran and surface conjugation with Alexa Fluor® 488 (AF488). Then, we investigated their intracellular distribution in HeLa cells over time. RESULTS The liposomes had similar physical properties and did not cause significant cell mortality after treatment for 180 min. The delivery rate and efficiency of encapsulated FITC-dextran with the fusogenic K3C16 liposomes were 3 and 1.6 times higher, respectively, than with the endocytic K3C14 liposomes. FITC-dextran molecules delivered with K3C16 liposomes were observed throughout the cytosolic space after 10 min, while those delivered with K3C14 liposomes were mainly observed as foci and took 60 min to diffuse into the cytosolic space. K3C14 lipids modified with AF488 were distributed mostly in the cytosolic space. In contrast, fluorescently labeled K3C16 lipids were colocalized with the plasma membrane of 50% of the HeLa cells after 10 min and were gradually internalized intracellularly. CONCLUSION Fusogenic K3C16 liposomes internalized into HeLa cells faster than endocytic K3C14 liposomes, and their components differently distributed in the cells.
Collapse
Affiliation(s)
- Masato Takikawa
- Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo169-8555, Japan
| | - Mizuki Fujisawa
- Department of Life Science and Medical Bioscience, Graduate Schoolof Advanced Science and Engineering, Waseda University (TWIns), Tokyo162-8480, Japan
| | - Kazuma Yoshino
- Department of Life Science and Medical Bioscience, Graduate Schoolof Advanced Science and Engineering, Waseda University (TWIns), Tokyo162-8480, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate Schoolof Advanced Science and Engineering, Waseda University (TWIns), Tokyo162-8480, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Tokyo169-8555, Japan
| |
Collapse
|
45
|
Shi J, Zhao D, Li X, Ding F, Tang X, Liu N, Huang H, Liu C. The conjugation of rhodamine B enables carrier-free mitochondrial delivery of functional proteins. Org Biomol Chem 2020; 18:6829-6839. [PMID: 32761021 DOI: 10.1039/d0ob01305f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
The development of protein-based therapeutics faces many challenges, for example, carrier-dependence, safety concerns, endocytosis-dependence, and uncertain in vivo therapeutic outcomes. Small molecules are rarely used for intracellular organelle-targeting and disease tissue-specific carrier-independent delivery of therapeutic proteins. Here, we report that rhodamine B, after modification with proteins, is able to guide carrier-free delivery into mitochondria and tissue-dependent distributions of functional proteins through organic cation transporters (OCTs). The enrichment of the modified catalase in the cancer tissue efficiently suppresses xenograft human lung tumor in mice. This carrier-free delivery platform of proteins may emerge as a simple yet powerful approach for cancer treatment.
Collapse
Affiliation(s)
- Jiayuan Shi
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensing Technology and Health, School of Chemistry, Central China Normal University, Wuhan, 430079 China.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Jin J, Wu Y, Chen J, Shen Y, Zhang L, Zhang H, Chen L, Yuan H, Chen H, Zhang W, Luan X. The peptide PROTAC modality: a novel strategy for targeted protein ubiquitination. Theranostics 2020; 10:10141-10153. [PMID: 32929339 PMCID: PMC7481416 DOI: 10.7150/thno.46985] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Despite dramatic advances in drug discovery over the decades, effective therapeutic strategies for cancers treatment are still in urgent demands. PROteolysis TArgeting Chimera (PROTAC), a novel therapeutic modality, has been vigorously promoted in preclinical and clinical applications. Unlike small molecule PROTAC, peptide PROTAC (p-PROTAC) with advantages of high specificity and low toxicity, while avoiding the limitations of shallow binding pockets through large interacting surfaces, provides promising substitutions for E3 ubiquitin ligase complex-mediated ubiquitination of "undruggable proteins". It is worth noting that successful applications of p-PROTAC still have some obstacles, including low stability and poor membrane permeability. Hence, we highlight that p-PROTAC combined with cell-penetrating peptides, constrained conformation technique, and targeted delivery systems could be the future efforts for potential translational research.
Collapse
Affiliation(s)
- Jinmei Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ye Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinjiao Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yiwen Shen
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lijun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lili Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hebao Yuan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109 US
| | - Hongzhuan Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
47
|
Knox SL, Steinauer A, Alpha-Cobb G, Trexler A, Rhoades E, Schepartz A. Quantification of protein delivery in live cells using fluorescence correlation spectroscopy. Methods Enzymol 2020; 641:477-505. [PMID: 32713536 DOI: 10.1016/bs.mie.2020.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fluorescence correlation spectroscopy (FCS) is a quantitative single-molecule method that measures the concentration and rate of diffusion of fluorophore-tagged molecules, both large and small, in vitro and within live cells, and even within discrete cellular compartments. FCS is exceptionally well-suited to directly quantify the efficiency of intracellular protein delivery-specifically, how well different "cell-penetrating" proteins and peptides guide proteinaceous materials into the cytosol and nuclei of live mammalian cells. This article provides an overview of the procedures necessary to execute robust FCS experiments and evaluate endosomal escape efficiencies: preparation of fluorophore-tagged proteins, incubation with mammalian cells and preparation of FCS samples, setup and execution of an FCS experiment, and a detailed discussion of and custom MATLAB® script for analyzing the resulting autocorrelation curves in the context of appropriate diffusion models.
Collapse
Affiliation(s)
- Susan L Knox
- Department of Chemistry, University of California, Berkeley, CA, United States
| | - Angela Steinauer
- Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Garrett Alpha-Cobb
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, United States
| | - Adam Trexler
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, United States
| | - Elizabeth Rhoades
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, United States
| | - Alanna Schepartz
- Department of Chemistry, University of California, Berkeley, CA, United States; Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States.
| |
Collapse
|
48
|
Ouyang L, Armstrong JPK, Chen Q, Lin Y, Stevens MM. Void-free 3D Bioprinting for In-situ Endothelialization and Microfluidic Perfusion. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909009. [PMID: 35677899 PMCID: PMC7612826 DOI: 10.1002/adfm.201909009] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Two major challenges of 3D bioprinting are the retention of structural fidelity and efficient endothelialization for tissue vascularization. We address both of these issues by introducing a versatile 3D bioprinting strategy, in which a templating bioink is deposited layer-by-layer alongside a matrix bioink to establish void-free multimaterial structures. After crosslinking the matrix phase, the templating phase is sacrificed to create a well-defined 3D network of interconnected tubular channels. This void-free 3D printing (VF-3DP) approach circumvents the traditional concerns of structural collapse, deformation and oxygen inhibition, moreover, it can be readily used to print materials that are widely considered "unprintable". By pre-loading endothelial cells into the templating bioink, the inner surface of the channels can be efficiently cellularized with a confluent endothelial layer. This in-situ endothelialization method can be used to produce endothelium with a far greater uniformity than can be achieved using the conventional post-seeding approach. This VF-3DP approach can also be extended beyond tissue fabrication and towards customized hydrogel-based microfluidics and self-supported perfusable hydrogel constructs.
Collapse
|
49
|
Kardani K, Bolhassani A, Agi E, Hashemi A. B1 protein: a novel cell penetrating protein for in vitro and in vivo delivery of HIV-1 multi-epitope DNA constructs. Biotechnol Lett 2020; 42:1847-1863. [PMID: 32449070 PMCID: PMC7246087 DOI: 10.1007/s10529-020-02918-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/17/2020] [Indexed: 12/02/2022]
Abstract
Objectives Enhancement of the potential ability of biomacromolecules to cross cell membranes is a critical step for development of effective therapeutic vaccine especially DNA vaccine against human immunodeficiency virus-1 (HIV-1) infection. The supercharged proteins were known as powerful weapons for delivery of different types of cargoes such as DNA and protein. Hence, we applied B1 protein with + 43 net charges obtained from a single frameshift in the gene encoding enhanced green fluorescent protein (eGFP) for delivery of two multi-epitope DNA constructs (nef-vpu-gp160-p24 and nef-vif-gp160-p24) in vitro and in vivo for the first time. For this purpose, B1 protein was generated in bacterial expression system under native conditions, and used to interact with both DNA constructs. Results Our data indicated that B1 protein (~ 27 kDa) was able to form a stable nanoparticle (~ 80–110 nm) with both DNA constructs at nitrogen: phosphate (N: P) ratio of 1:1. Moreover, the transfection efficiency of B1 protein for DNA delivery into HEK-293T cell line indicated that the cellular uptake of nef-vif-gp160-p24 DNA/ B1 and nef-vpu-gp160-p24 DNA/ B1 nanoparticles was about 32–35% with lower intensity as compared to TurboFect commercial reagent. On the other hand, immunization of BALB/c mice with different modalities demonstrated that B1 protein could enhance the levels of antibody, IFN-gamma and Granzyme B eliciting potent and strong Th1-directed cellular immunity. Conclusion Generally, our findings showed the potency of B1 protein as a promising gene delivery system to improve an effective therapeutic vaccine against HIV-1 infection.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Elnaz Agi
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Atieh Hashemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
A myosin-7B-dependent endocytosis pathway mediates cellular entry of α-synuclein fibrils and polycation-bearing cargos. Proc Natl Acad Sci U S A 2020; 117:10865-10875. [PMID: 32366666 DOI: 10.1073/pnas.1918617117] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cell-to-cell transmission of misfolding-prone α-synuclein (α-Syn) has emerged as a key pathological event in Parkinson's disease. This process is initiated when α-Syn-bearing fibrils enter cells via clathrin-mediated endocytosis, but the underlying mechanisms are unclear. Using a CRISPR-mediated knockout screen, we identify SLC35B2 and myosin-7B (MYO7B) as critical endocytosis regulators for α-Syn preformed fibrils (PFFs). We show that SLC35B2, as a key regulator of heparan sulfate proteoglycan (HSPG) biosynthesis, is essential for recruiting α-Syn PFFs to the cell surface because this process is mediated by interactions between negatively charged sugar moieties of HSPGs and clustered K-T-K motifs in α-Syn PFFs. By contrast, MYO7B regulates α-Syn PFF cell entry by maintaining a plasma membrane-associated actin network that controls membrane dynamics. Without MYO7B or actin filaments, many clathrin-coated pits fail to be severed from the membrane, causing accumulation of large clathrin-containing "scars" on the cell surface. Intriguingly, the requirement for MYO7B in endocytosis is restricted to α-Syn PFFs and other polycation-bearing cargos that enter cells via HSPGs. Thus, our study not only defines regulatory factors for α-Syn PFF endocytosis, but also reveals a previously unknown endocytosis mechanism for HSPG-binding cargos in general, which requires forces generated by MYO7B and actin filaments.
Collapse
|