1
|
He W, Yang F, Chen K, Zeng Q. Targeted gold nanoparticles for ovarian cancer (Review). Oncol Lett 2024; 28:589. [PMID: 39417039 PMCID: PMC11481100 DOI: 10.3892/ol.2024.14723] [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: 06/07/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Among all malignant gynecological tumors, ovarian cancer (OC) has the highest mortality rate. OC is often diagnosed at advanced and incurable stages; however, early diagnosis can enable the use of optimized and personalized treatments. Intensive research into the synthesis and characterization of gold nanoparticles (AuNPs) has been performed with the aim of developing innovative materials for use in biological and photothermal therapies for OC. AuNPs can be chemically modified and functionalized by binding to a variety of organic compounds and biomolecules, such as peptides, antibodies and therapeutic agents, via simple synthetic processes. They are particularly suitable for use as carriers for drug delivery. In the present review, the synthesis and characteristics of AuNPs are summarized, and their potential in OC therapy are discussed.
Collapse
Affiliation(s)
- Wenjuan He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Fuyuan Yang
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, Hubei 434000, P.R. China
| | - Keming Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Qingsong Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
2
|
Chen K, Liu Y, Duan G, Shi M, Yang C, Xing R, Yan X. Biomolecular Condensates Based on Amino Acid for Enhancing Oral Bioavailability and Therapeutic Efficacy of Hydrophobic Drugs. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58370-58378. [PMID: 39404746 DOI: 10.1021/acsami.4c13792] [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: 11/01/2024]
Abstract
The oral administration of chemo- or immunotherapeutic drugs presents a compelling alternative for patients with malignant colorectal cancer, offering a convenient and patient-compliant "hospital-free" strategy. Unfortunately, the hydrophobic nature of many drug candidates, alongside the harsh conditions of the gastrointestinal tract, frequently results in suboptimal bioavailability and heightened systemic toxicity. To address these challenges, we harnessed the unique properties of biomolecular condensates, which form through a liquid-liquid phase separation mechanism, to develop a versatile platform for drug encapsulation and delivery. In this study, we introduce a reliable and effective amorphous oral drug delivery system based on biomolecular condensates derived from the amino acid derivative N-(benzyloxycarbonyl)-l-proline (ZP). These ZP condensates exhibit dynamic intermolecular interactions and possess unique physicochemical attributes such as fluidity and viscoelasticity. They significantly improve the solubility of hydrophobic drugs, ensuring enhanced stability and optimized pharmacokinetics under physiological and gastrointestinal conditions. By maintaining drugs in an amorphous state, we substantially increased drug bioavailability and markedly improved pharmacokinetics. Furthermore, the ZP condensates demonstrate potential as an integrated therapeutic platform capable of potentiating the synergies between chemotherapy and immunotherapy while concurrently reducing systemic toxicity. This has resulted in a significant enhancement of chemo-immunotherapy efficacy in the treatment of colorectal cancer, representing a notable advancement in drug delivery and oncology.
Collapse
Affiliation(s)
- Kaiwei Chen
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yazhou Liu
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Guifang Duan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Mengqian Shi
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Chaojuan Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100083, China
| | - Ruirui Xing
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Center for Mesoscience, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
3
|
Ma X, Zhang Z, Barba-Bon A, Han D, Qi Z, Ge B, He H, Huang F, Nau WM, Wang X. A small-molecule carrier for the intracellular delivery of a membrane-impermeable protein with retained bioactivity. Proc Natl Acad Sci U S A 2024; 121:e2407515121. [PMID: 39436658 PMCID: PMC11536097 DOI: 10.1073/pnas.2407515121] [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: 04/15/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Intracellular protein delivery has the potential to revolutionize cell-biological research and medicinal therapy, with broad applications in bioimaging, disease treatment, and genome editing. Herein, we demonstrate successful delivery of a functional protein, cytochrome c (CYC), by using a boron cluster anion as molecular carrier of the superchaotropic anion type (B12Br11OPr2-). CYC was delivered into lipid bilayer vesicles as well as living cells, with a cellular uptake ratio approaching 90%. Mechanistic studies showed that CYC was internalized into cells through a permeation pathway directly into the cytoplasm, bypassing endosomal entrapment. Upon carrier-assisted internalization, CYC retained its bioactivity, as reflected by an induced cell apoptosis rate of 25% at low dose (1 µM). This study furbishes a direct protein delivery method by a molecular carrier with high efficiency, confirming the potential of inorganic cluster ions as protein transport vehicles with an extensive range of future cell-biological or biomedical applications.
Collapse
Affiliation(s)
- Xiqi Ma
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Zhixiong Zhang
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | | | - Dongxue Han
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Zichun Qi
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Baosheng Ge
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Hua He
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Fang Huang
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Werner M. Nau
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
- School of Science, Constructor University, Bremen28759, Germany
| | - Xiaojuan Wang
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| |
Collapse
|
4
|
Su H, Rong G, Li L, Cheng Y. Subcellular targeting strategies for protein and peptide delivery. Adv Drug Deliv Rev 2024; 212:115387. [PMID: 38964543 DOI: 10.1016/j.addr.2024.115387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/15/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Cytosolic delivery of proteins and peptides provides opportunities for effective disease treatment, as they can specifically modulate intracellular processes. However, most of protein-based therapeutics only have extracellular targets and are cell-membrane impermeable due to relatively large size and hydrophilicity. The use of organelle-targeting strategy offers great potential to overcome extracellular and cell membrane barriers, and enables localization of protein and peptide therapeutics in the organelles. Although progresses have been made in the recent years, organelle-targeted protein and peptide delivery is still challenging and under exploration. We reviewed recent advances in subcellular targeted delivery of proteins/peptides with a focus on targeting mechanisms and strategies, and highlight recent examples of active and passive organelle-specific protein and peptide delivery systems. This emerging platform could open a new avenue to develop more effective protein and peptide therapeutics.
Collapse
Affiliation(s)
- Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Guangyu Rong
- Department of Ophthalmology and Vision Science, Shanghai Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, 200030, China
| | - Longjie Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
5
|
Mixová G, Tihlaříková E, Zhu Y, Schindler L, Androvič L, Kracíková L, Hrdá E, Porsch B, Pechar M, Garliss CM, Wilson D, Welles HC, Holechek J, Ren Q, Lynn GM, Neděla V, Laga R. Synthesis and Structure Optimization of Star Copolymers as Tunable Macromolecular Carriers for Minimal Immunogen Vaccine Delivery. Bioconjug Chem 2024; 35:1218-1232. [PMID: 39081220 PMCID: PMC11342300 DOI: 10.1021/acs.bioconjchem.4c00273] [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: 06/14/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Minimal immunogen vaccines are being developed to focus antibody responses against otherwise challenging targets, including human immunodeficiency virus (HIV), but multimerization of the minimal peptide immunogen on a carrier platform is required for activity. Star copolymers comprising multiple hydrophilic polymer chains ("arms") radiating from a central dendrimer unit ("core") were recently reported to be an effective platform for arraying minimal immunogens for inducing antibody responses in mice and primates. However, the impact of different parameters of the star copolymer (e.g., minimal immunogen density and hydrodynamic size) on antibody responses and the optimal synthetic route for controlling those parameters remains to be fully explored. We synthesized a library of star copolymers composed of poly[N-(2-hydroxypropyl)methacrylamide] hydrophilic arms extending from poly(amidoamine) dendrimer cores with the aim of identifying the optimal composition for use as minimal immunogen vaccines. Our results show that the length of the polymer arms has a crucial impact on the star copolymer hydrodynamic size and is precisely tunable over a range of 20-50 nm diameter, while the dendrimer generation affects the maximum number of arms (and therefore minimal immunogens) that can be attached to the surface of the dendrimer. In addition, high-resolution images of selected star copolymer taken by a custom-modified environmental scanning electron microscope enabled the acquisition of high-resolution images, providing new insights into the star copolymer structure. Finally, in vivo studies assessing a star copolymer vaccine comprising an HIV minimal immunogen showed the criticality of polymer arm length in promoting antibody responses and highlighting the importance of composition tunability to yield the desired biological effect.
Collapse
Affiliation(s)
- Gabriela Mixová
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Eva Tihlaříková
- Institute
of Scientific Instruments, Czech Academy
of Sciences, Královopolská
147, Brno 612 64, Czech Republic
| | - Yaling Zhu
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Lucie Schindler
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Ladislav Androvič
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Lucie Kracíková
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Eliška Hrdá
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Bedřich Porsch
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Michal Pechar
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| | - Christopher M. Garliss
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - David Wilson
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Hugh C. Welles
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Jake Holechek
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Qiuyin Ren
- Vaccine
Research Center, National Institutes of
Health, Rockville, Maryland 20892, United States
| | - Geoffrey M. Lynn
- Barinthus
Biotherapeutics North America, Inc. (formerly Avidea Technologies,
Inc.), 20400 Century
Boulevard, Germantown, Maryland 20874, United States
| | - Vilém Neděla
- Institute
of Scientific Instruments, Czech Academy
of Sciences, Královopolská
147, Brno 612 64, Czech Republic
| | - Richard Laga
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
nám. 2, Prague 162
06, Czech Republic
| |
Collapse
|
6
|
Qutub SS, Bhat IA, Maatouk BI, Moosa B, Fakim A, Nawaz K, Diaz-Galicia E, Lin W, Grünberg R, Arold ST, Khashab NM. An Amphiphilic Cell-Penetrating Macrocycle for Efficient Cytosolic Delivery of Proteins, DNA, and CRISPR Cas9. Angew Chem Int Ed Engl 2024; 63:e202403647. [PMID: 38752721 DOI: 10.1002/anie.202403647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Indexed: 07/02/2024]
Abstract
The discovery of safe platforms that can circumvent the endocytic pathway is of great significance for biological therapeutics that are usually degraded during endocytosis. Here we show that a self-assembled and dynamic macrocycle can passively diffuse through the cell membrane and deliver a broad range of biologics, including proteins, CRISPR Cas9, and ssDNA, directly to the cytosol while retaining their bioactivity. Cell-penetrating macrocycle CPM can be easily prepared from the room temperature condensation of diketopyrrolopyrrole lactams with diamines. We attribute the high cellular permeability of CPM to its amphiphilic nature and chameleonic properties. It adopts conformations that partially bury polar groups and expose hydrophobic side chains, thus self-assembling into micellar-like structures. Its superior fluorescence makes CPM trackable inside cells where it follows the endomembrane system. CPM outperformed commercial reagents for biologics delivery and showed high RNA knockdown efficiency of CRISPR Cas9. We envisage that this macrocycle will be an ideal starting point to design and synthesize biomimetic macrocyclic tags that can readily facilitate the interaction and uptake of biomolecules and overcome endosomal digestion.
Collapse
Affiliation(s)
- Somayah S Qutub
- Smart Hybrid Materials (SHMs) Laboratory, Chemistry Program, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Imtiyaz Ahmad Bhat
- Smart Hybrid Materials (SHMs) Laboratory, Chemistry Program, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Current Address: Department of Chemistry, Islamic University of Science and Technology, Awantipora, 192122, Jammu and Kashmir, India
| | - Batoul I Maatouk
- Smart Hybrid Materials (SHMs) Laboratory, Chemistry Program, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Basem Moosa
- Smart Hybrid Materials (SHMs) Laboratory, Chemistry Program, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Aliyah Fakim
- Smart Hybrid Materials (SHMs) Laboratory, Chemistry Program, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Kashif Nawaz
- The Coral Symbiomics Lab, Red Sea Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Escarlet Diaz-Galicia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Weibin Lin
- Smart Hybrid Materials (SHMs) Laboratory, Chemistry Program, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Raik Grünberg
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Stefan T Arold
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Niveen M Khashab
- Smart Hybrid Materials (SHMs) Laboratory, Chemistry Program, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| |
Collapse
|
7
|
Agrohia DK, Goswami R, Jantarat T, Çiçek YA, Thongsukh K, Jeon T, Bell JM, Rotello VM, Vachet RW. Suborgan Level Quantitation of Proteins in Tissues Delivered by Polymeric Nanocarriers. ACS NANO 2024; 18:16808-16818. [PMID: 38870478 PMCID: PMC11497159 DOI: 10.1021/acsnano.4c02344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Amidst the rapid growth of protein therapeutics as a drug class, there is an increased focus on designing systems to effectively deliver proteins to target organs. Quantitative monitoring of protein distributions in tissues is essential for optimal development of delivery systems; however, existing strategies can have limited accuracy, making it difficult to assess suborgan dosing. Here, we describe a quantitative imaging approach that utilizes metal-coded mass tags and laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) to quantify the suborgan distributions of proteins in tissues that have been delivered by polymeric nanocarriers. Using this approach, we measure nanomole per gram levels of proteins as delivered by guanidinium-functionalized poly(oxanorborneneimide) (PONI) polymers to various tissues, including the alveolar region of the lung. Due to the multiplexing capability of the LA-ICP-MS imaging, we are also able to simultaneously quantify protein and polymer distributions, obtaining valuable information about the relative excretion pathways of the protein cargo and carrier. This imaging approach will facilitate quantitative correlations between nanocarrier properties and protein cargo biodistributions.
Collapse
Affiliation(s)
- Dheeraj K. Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Teerapong Jantarat
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Yağız Anil Çiçek
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Korndanai Thongsukh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jonathan M. Bell
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
8
|
Behzadipour Y, Hemmati S. Covalent conjugation and non-covalent complexation strategies for intracellular delivery of proteins using cell-penetrating peptides. Biomed Pharmacother 2024; 176:116910. [PMID: 38852512 DOI: 10.1016/j.biopha.2024.116910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Therapeutic proteins provided new opportunities for patients and high sales volumes. However, they are formulated for extracellular targets. The lipophilic barrier of the plasma membrane renders the vast array of intracellular targets out of reach. Peptide-based delivery systems, namely cell-penetrating peptides (CPPs), have few safety concerns, and low immunogenicity, with control over administered doses. This study investigates CPP-based protein delivery systems by classifying them into CPP-protein "covalent conjugation" and CPP: protein "non-covalent complexation" categories. Covalent conjugates ensure the proximity of the CPP to the cargo, which can improve cellular uptake and endosomal escape. We will discuss various aspects of covalent conjugates through non-cleavable (stable) or cleavable bonds. Non-cleavable CPP-protein conjugates are produced by recombinant DNA technology to express the complete fusion protein in a host cell or by chemical ligation of CPP and protein, which ensures stability during the delivery process. CPP-protein cleavable bonds are classified into pH-sensitive and redox-sensitive bonds, enzyme-cleavable bonds, and physical stimuli cleavable linkers (light radiation, ultrasonic waves, and thermo-responsive). We have highlighted the key characteristics of non-covalent complexes through electrostatic and hydrophobic interactions to preserve the conformational integrity of the CPP and cargo. CPP-mediated protein delivery by non-covalent complexation, such as zippers, CPP adaptor methods, and avidin-biotin technology, are featured. Conclusively, non-covalent complexation methods are appropriate when a high number of CPP or protein samples are to be screened. In contrast, when the high biological activity of the protein is critical in the intracellular compartment, conjugation protocols are preferred.
Collapse
Affiliation(s)
- Yasaman Behzadipour
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran
| | - Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran; Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran.
| |
Collapse
|
9
|
Ling QH, Lou ZC, Zhang L, Jin T, Dou WT, Yang HB, Xu L. Supramolecular cage-mediated cargo transport. Chem Soc Rev 2024; 53:6042-6067. [PMID: 38770558 DOI: 10.1039/d3cs01081c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
A steady stream of material transport based on carriers and channels in living systems plays an extremely important role in normal life activities. Inspired by nature, researchers have extensively applied supramolecular cages in cargo transport because of their unique three-dimensional structures and excellent physicochemical properties. In this review, we will focus on the development of supramolecular cages as carriers and channels for cargo transport in abiotic and biological systems over the past fifteen years. In addition, we will discuss future challenges and potential applications of supramolecular cages in substance transport.
Collapse
Affiliation(s)
- Qing-Hui Ling
- State Key Laboratory of Petroleum Molecular and Process Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Wuhu Hospital Affiliated to East China Normal University (The Second People's Hospital of Wuhu), Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200241, China.
| | - Zhen-Chen Lou
- State Key Laboratory of Petroleum Molecular and Process Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Wuhu Hospital Affiliated to East China Normal University (The Second People's Hospital of Wuhu), Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200241, China.
| | - Lei Zhang
- Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, China
| | - Tongxia Jin
- State Key Laboratory of Petroleum Molecular and Process Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Wuhu Hospital Affiliated to East China Normal University (The Second People's Hospital of Wuhu), Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200241, China.
| | - Wei-Tao Dou
- State Key Laboratory of Petroleum Molecular and Process Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Wuhu Hospital Affiliated to East China Normal University (The Second People's Hospital of Wuhu), Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200241, China.
| | - Hai-Bo Yang
- State Key Laboratory of Petroleum Molecular and Process Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Wuhu Hospital Affiliated to East China Normal University (The Second People's Hospital of Wuhu), Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200241, China.
| | - Lin Xu
- State Key Laboratory of Petroleum Molecular and Process Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, Wuhu Hospital Affiliated to East China Normal University (The Second People's Hospital of Wuhu), Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Road, Shanghai 200241, China.
| |
Collapse
|
10
|
Song J, Fransen PPKH, Bakker MH, Wijnands SPW, Huang J, Guo S, Dankers PYW. The effect of charge and albumin on cellular uptake of supramolecular polymer nanostructures. J Mater Chem B 2024; 12:4854-4866. [PMID: 38682307 PMCID: PMC11111113 DOI: 10.1039/d3tb02631k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/30/2024] [Indexed: 05/01/2024]
Abstract
Intracellular delivery of functional biomolecules by using supramolecular polymer nanostructures has gained significant interest. Here, various charged supramolecular ureido-pyrimidinone (UPy)-aggregates were designed and formulated via a simple "mix-and-match" method. The cellular internalization of these UPy-aggregates in the presence or absence of serum proteins by phagocytic and non-phagocytic cells, i.e., THP-1 derived macrophages and immortalized human kidney cells (HK-2 cells), was systematically investigated. In the presence of serum proteins the UPy-aggregates were taken up by both types of cells irrespective of the charge properties of the UPy-aggregates, and the UPy-aggregates co-localized with mitochondria of the cells. In the absence of serum proteins only cationic UPy-aggregates could be effectively internalized by THP-1 derived macrophages, and the internalized UPy-aggregates either co-localized with mitochondria or displayed as vesicular structures. While the cationic UPy-aggregates were hardly internalized by HK-2 cells and could only bind to the membrane of HK-2 cells. With adding and increasing the amount of serum albumin in the cell culture medium, the cationic UPy-aggregates were gradually taken up by HK-2 cells without anchoring on the cell membranes. It is proposed that the serum albumin regulates the cellular internalization of UPy-aggregates. These results provide fundamental insights for the fabrication of supramolecular polymer nanostructures for intracellular delivery of therapeutics.
Collapse
Affiliation(s)
- Jiankang Song
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory for Cell and Tissue Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Peter-Paul K H Fransen
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Maarten H Bakker
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Sjors P W Wijnands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Jingyi Huang
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Shuaiqi Guo
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
| | - Patricia Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory for Cell and Tissue Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| |
Collapse
|
11
|
Kong H, Zheng C, Yi K, Mintz RL, Lao YH, Tao Y, Li M. An antifouling membrane-fusogenic liposome for effective intracellular delivery in vivo. Nat Commun 2024; 15:4267. [PMID: 38769317 PMCID: PMC11106281 DOI: 10.1038/s41467-024-46533-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 03/01/2024] [Indexed: 05/22/2024] Open
Abstract
The membrane-fusion-based internalization without lysosomal entrapment is advantageous for intracellular delivery over endocytosis. However, protein corona formed on the membrane-fusogenic liposome surface converts its membrane-fusion performance to lysosome-dependent endocytosis, causing poorer delivery efficiency in biological conditions. Herein, we develop an antifouling membrane-fusogenic liposome for effective intracellular delivery in vivo. Leveraging specific lipid composition at an optimized ratio, such antifouling membrane-fusogenic liposome facilitates fusion capacity even in protein-rich conditions, attributed to the copious zwitterionic phosphorylcholine groups for protein-adsorption resistance. Consequently, the antifouling membrane-fusogenic liposome demonstrates robust membrane-fusion-mediated delivery in the medium with up to 38% fetal bovine serum, outclassing two traditional membrane-fusogenic liposomes effective at 4% and 6% concentrations. When injected into mice, antifouling membrane-fusogenic liposomes can keep their membrane-fusion-transportation behaviors, thereby achieving efficient luciferase transfection and enhancing gene-editing-mediated viral inhibition. This study provides a promising tool for effective intracellular delivery under complex physiological environments, enlightening future nanomedicine design.
Collapse
Affiliation(s)
- Huimin Kong
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- School of Chemistry, South China Normal University, Guangzhou, 510006, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Rachel L Mintz
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
12
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
13
|
Wang B, Xu XJ, Fu Y, Ren B, Yang XD, Yang HY. A tumor-targeted and enzyme-responsive gold nanorod-based nanoplatform with facilitated endo-lysosomal escape for synergetic photothermal therapy and protein therapy. Dalton Trans 2024; 53:2120-2130. [PMID: 38180436 DOI: 10.1039/d3dt03305h] [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: 01/06/2024]
Abstract
To tackle the obstacles related to tumor targeting and overcome the limitations of single treatment models, we have developed a nanoplatform that is both tumor-targeted and enzyme-responsive. This nanoplatform integrates photothermal gold nanorods (AuNRs) and protein drugs into a single system. This nanosystem, known as AuNRs@HA-mPEG-Deta-LA, was fabricated by modifying gold nanorods (AuNRs) with a polymeric ligand called hyaluronic acid-grafted-(mPEG/diethylenetriamine-conjugated-lipoic acid). The purpose of this fabrication was to load cytochrome c (CC) and utilize it for the synergetic protein-photothermal therapy of cancer. The resulting nanoplatform exhibited a high efficiency in loading proteins and demonstrated excellent stability in different biological environments. Additionally, CC-loaded AuNRs@HA-mPEG-Deta-LA not only enabled localized hyperthermia for photothermal therapy (PTT) with laser irradiation but also facilitated the release of CC under the action of hyaluronidase, an enzyme known to be overexpressed in tumor cells. The confocal imaging results demonstrated that the presence of a specific polymeric ligand on this nanoparticle enhances the internalization of CD44-positive cancer cells, accelerates endo/lysosomal escape, and facilitates the controlled release of CC within the cells. Furthermore, the results of the MTT assay also showed that AuNRs@HA-mPEG-Deta-LA as a protein nanocarrier demonstrated excellent biocompatibility. Importantly, this synergistic therapeutic strategy effectively induced apoptosis in A549 cancer cells by increasing the intracellular concentration of CC and utilizing the photothermal conversion of AuNRs, which was observed to be more effective compared to using only protein therapy or PTT. Therefore, this study showcased a nanoplatform based on AuNRs that has great potential for tumor-targeted protein delivery in combination with PTT in cancer treatment.
Collapse
Affiliation(s)
- Bo Wang
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Xin Jun Xu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| | - Yan Fu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| | - Bo Ren
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Xiao Dong Yang
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| |
Collapse
|
14
|
Chan A, Tsourkas A. Intracellular Protein Delivery: Approaches, Challenges, and Clinical Applications. BME FRONTIERS 2024; 5:0035. [PMID: 38282957 PMCID: PMC10809898 DOI: 10.34133/bmef.0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/14/2023] [Indexed: 01/30/2024] Open
Abstract
Protein biologics are powerful therapeutic agents with diverse inhibitory and enzymatic functions. However, their clinical use has been limited to extracellular applications due to their inability to cross plasma membranes. Overcoming this physiological barrier would unlock the potential of protein drugs for the treatment of many intractable diseases. In this review, we highlight progress made toward achieving cytosolic delivery of recombinant proteins. We start by first considering intracellular protein delivery as a drug modality compared to existing Food and Drug Administration-approved drug modalities. Then, we summarize strategies that have been reported to achieve protein internalization. These techniques can be broadly classified into 3 categories: physical methods, direct protein engineering, and nanocarrier-mediated delivery. Finally, we highlight existing challenges for cytosolic protein delivery and offer an outlook for future advances.
Collapse
Affiliation(s)
| | - Andrew Tsourkas
- Department of Bioengineering,
University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
15
|
Xie F, Tang S, Zhang Y, Zhao Y, Lin Y, Yao Y, Wang M, Gu Z, Wan J. Designing Peptide-Based Nanoinhibitors of Programmed Cell Death Ligand 1 (PD-L1) for Enhanced Chemo-immunotherapy. ACS NANO 2024; 18:1690-1701. [PMID: 38165832 DOI: 10.1021/acsnano.3c09968] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
The combination of immune checkpoint blockade (ICB) and chemotherapy has shown significant potential in the clinical treatment of various cancers. However, circulating regeneration of PD-L1 within tumor cells greatly limits the efficiency of chemo-immunotherapy and consequent patient response rates. Herein, we report the synthesis of a nanoparticle-based PD-L1 inhibitor (FRS) with a rational design for effective endogenous PD-L1 suppression. The nanoinhibitor is achieved through self-assembly of fluoroalkylated competitive peptides that target PD-L1 palmitoylation. The FRS nanoparticles provide efficient protection and delivery of functional peptides to the cytoplasm of tumors, showing greater inhibition of PD-L1 than nonfluorinated peptidic inhibitors. Moreover, we demonstrate that FRS synergizes with chemotherapeutic doxorubicin (DOX) to boost the antitumor activities via simultaneous reduction of PD-L1 abundance and induction of immunogenic cell death in murine colon tumor models. The nano strategy of PD-L1 regulation present in this study is expected to advance the development of ICB inhibitors and overcome the limitations of conventional ICB-assisted chemo-immunotherapy.
Collapse
Affiliation(s)
- Fengjuan Xie
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
| | - Shasha Tang
- Department of Breast Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, People's Republic of China
| | - Ye Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
| | - Yinbing Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
| | - Yingying Lin
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
| | - Yining Yao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
| | - Meiyan Wang
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Zhengying Gu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
| |
Collapse
|
16
|
Chen X, Zheng Q, Cai W, Sheng J, Wang M. Biodegradable Hydrogen-Bonded Organic Framework for Cytosolic Protein Delivery. ACS APPLIED MATERIALS & INTERFACES 2023; 15:54346-54352. [PMID: 37967322 DOI: 10.1021/acsami.3c14450] [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: 11/17/2023]
Abstract
Hydrogen-bonded organic frameworks (HOFs) are a novel class of porous nanomaterials that show great potential for intracellular delivery of protein therapeutics. However, the inherent challenges in interfacing protein with HOFs, and the need for spatiotemporally controlling the release of protein within cells, have constrained their therapeutic potential. In this study, we report novel biodegradable hydrogen-bonded organic frameworks, termed DS-HOFs, specially designed for the cytosolic delivery of protein therapeutics in cancer cells. The synthesis of DS-HOFs involves the self-assembly of 4-[tris(4-carbamimidoylphenyl) methyl] benzenecarboximidamide (TAM) and 4,4'-dithiobisbenzoic acid (DTBA), governed by intermolecular hydrogen-bonding interactions. DS-HOFs exhibit high efficiency in encapsulating a diverse range of protein cargos, underpinned by the hydrogen-bonding interactions between the protein residue and DS-HOF subcomponents. Notably, DS-HOFs are selectively degraded in cancer cells triggered by the distinct intracellular reductive microenvironments, enabling an enhanced and selective release of protein inside cancer cells. Additionally, we demonstrate that the efficient delivery of bacterial effector protein DUF5 using DS-HOFs depletes the mutant RAS in cancer cells to prohibit tumor cell growth both in vitro and in vivo. The design of biodegradable HOFs for cytosolic protein delivery provides a powerful and promising strategy to expand the therapeutic potential of proteins for cancer therapy.
Collapse
Affiliation(s)
- Xianghan Chen
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qizhen Zheng
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiqi Cai
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinhan Sheng
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Wang
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Zhang S, Nakata E, Lin P, Morii T. An Artificial Liposome Compartment with Size Exclusion Molecular Transport. Chemistry 2023; 29:e202302093. [PMID: 37668304 DOI: 10.1002/chem.202302093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/28/2023] [Accepted: 09/04/2023] [Indexed: 09/06/2023]
Abstract
The cellular compartment plays an essential role in organizing the complex and diverse biochemical reactions within the cell. By mimicking the function of such cellular compartments, the challenge of constructing artificial compartments has been taken up to develop new biochemical tools for efficient material production and diagnostics. The important features required for the artificial compartment are that it isolates the interior from the external environment and is further functionalized to control the transport of target chemicals to regulate the interior concentration of both substrate and reaction products. In this study, an artificial compartment with size-selective molecular transport function was constructed by using a DNA origami-guided liposome prepared by modifying the method reported by Perrault et al. This completely isolates the liposome interior, including the DNA origami skeleton, from the external environment and allows the assembly of a defined number of molecules of interest inside and/or outside the compartment. By incorporating a bacterial membrane protein, OmpF, into the liposome, the resulting artificial compartment was shown to transport only the molecule of interest with a molecular weight below 600 Da from the external environment into the interior of the compartment.
Collapse
Affiliation(s)
- Shiwei Zhang
- Institute of Advanced Energy, Kyoto University Uji, Kyoto, 6110011, Japan
| | - Eiji Nakata
- Institute of Advanced Energy, Kyoto University Uji, Kyoto, 6110011, Japan
| | - Peng Lin
- Institute of Advanced Energy, Kyoto University Uji, Kyoto, 6110011, Japan
| | - Takashi Morii
- Institute of Advanced Energy, Kyoto University Uji, Kyoto, 6110011, Japan
| |
Collapse
|
18
|
Sim Y, Seong J, Lee S, Kim D, Choi E, Lah MS, Ryu JH. Metal-Organic Framework-Supported Catalase Delivery for Enhanced Photodynamic Therapy via Hypoxia Mitigation. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37883653 DOI: 10.1021/acsami.3c13395] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Tumor hypoxia poses a significant challenge in photodynamic therapy (PDT), which uses molecular oxygen to produce reactive oxygen species upon light excitation of a photosensitizer. For hypoxia mitigation, an enzyme catalase (CAT) can be beneficially used to convert intracellular hydrogen peroxide to molecular oxygen, but its utility is significantly limited due to the intrinsic membrane impermeability. Herein, we present direct integration of CAT into the outer surface of unmodified metal-organic framework (MOF) nanoparticles (NPs) via supramolecular interactions for effective cellular entry of CAT and consequent enhancement of PDT. The results demonstrated that CAT-loaded MOF NPs could successfully enter hypoxic cancer cells, after which the intracellularly delivered CAT molecules became dissociated from the MOF surface to efficiently initiate the oxygen generation and PDT process along with a co-delivered photosensitizer IR780. This achievement suggests that our protein-MOF integration strategy holds great potential in biomedical studies to overcome tumor hypoxia as well as to efficiently deliver biomolecular cargos.
Collapse
Affiliation(s)
- Youjung Sim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Junmo Seong
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seonghwan Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Dohyun Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eunshil Choi
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Myoung Soo Lah
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
19
|
Wolff N, Loza K, Heggen M, Schaller T, Niemeyer F, Bayer P, Beuck C, Oliveira CLP, Prymak O, Weidenthaler C, Epple M. Ultrastructure and Surface Composition of Glutathione-Terminated Ultrasmall Silver, Gold, Platinum, and Alloyed Silver-Platinum Nanoparticles (2 nm). Inorg Chem 2023; 62:17470-17485. [PMID: 37820300 DOI: 10.1021/acs.inorgchem.3c02879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Alloyed ultrasmall silver-platinum nanoparticles (molar ratio Ag:Pt = 50:50) were prepared and compared to pure silver, platinum, and gold nanoparticles, all with a metallic core diameter of 2 nm. They were surface-stabilized by a layer of glutathione (GSH). A comprehensive characterization by high-resolution transmission electron microscopy (HRTEM), electron diffraction (ED), X-ray diffraction (XRD), small-angle X-ray scattering (SAXS), differential centrifugal sedimentation (DCS), and UV spectroscopy showed their size both in the dry and in the water-dispersed state (hydrodynamic diameter). Solution NMR spectroscopy (1H, 13C, COSY, HSQC, HMBC, and DOSY) showed the nature of the glutathione shell including the number of GSH ligands on each nanoparticle (about 200 with a molecular footprint of 0.063 nm2 each). It furthermore showed that there are at least two different positions for the GSH ligand on the gold nanoparticle surface. Platinum strongly reduced the resolution of the NMR spectra compared to silver and gold, also in the alloyed nanoparticles. X-ray photoelectron spectroscopy (XPS) showed that silver, platinum, and silver-platinum particles were at least partially oxidized to Ag(+I) and Pt(+II), whereas the gold nanoparticles showed no sign of oxidation. Platinum and gold nanoparticles were well crystalline but twinned (fcc lattice) despite the small particle size. Silver was crystalline in electron diffraction but not in X-ray diffraction. Alloyed silver-platinum nanoparticles were almost fully amorphous by both methods, indicating a considerable internal disorder.
Collapse
Affiliation(s)
- Natalie Wolff
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Essen 45117, Germany
| | - Kateryna Loza
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Essen 45117, Germany
| | - Marc Heggen
- Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, Forschungszentrum Jülich GmbH, Jülich 52428, Germany
| | - Torsten Schaller
- Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Felix Niemeyer
- Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Peter Bayer
- Structural and Medicinal Biochemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Christine Beuck
- Structural and Medicinal Biochemistry, University of Duisburg-Essen, Essen 45117, Germany
| | | | - Oleg Prymak
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Essen 45117, Germany
| | - Claudia Weidenthaler
- Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, Mülheim an der Ruhr 45470, Germany
| | - Matthias Epple
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Essen 45117, Germany
| |
Collapse
|
20
|
Trimaille T, Verrier B. Copolymer Micelles: A Focus on Recent Advances for Stimulus-Responsive Delivery of Proteins and Peptides. Pharmaceutics 2023; 15:2481. [PMID: 37896241 PMCID: PMC10609739 DOI: 10.3390/pharmaceutics15102481] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Historically used for the delivery of hydrophobic drugs through core encapsulation, amphiphilic copolymer micelles have also more recently appeared as potent nano-systems to deliver protein and peptide therapeutics. In addition to ease and reproducibility of preparation, micelles are chemically versatile as hydrophobic/hydrophilic segments can be tuned to afford protein immobilization through different approaches, including non-covalent interactions (e.g., electrostatic, hydrophobic) and covalent conjugation, while generally maintaining protein biological activity. Similar to many other drugs, protein/peptide delivery is increasingly focused on stimuli-responsive nano-systems able to afford triggered and controlled release in time and space, thereby improving therapeutic efficacy and limiting side effects. This short review discusses advances in the design of such micelles over the past decade, with an emphasis on stimuli-responsive properties for optimized protein/peptide delivery.
Collapse
Affiliation(s)
- Thomas Trimaille
- Ingénierie des Matériaux Polymères, Univ Lyon, CNRS, Université Claude Bernard Lyon 1, INSA Lyon, Université Jean Monnet, UMR 5223, CEDEX, 69622 Villeurbanne, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Univ Lyon, CNRS, Université Claude Bernard Lyon 1, UMR 5305, 7 Passage du Vercors, CEDEX 07, 69367 Lyon, France;
| |
Collapse
|
21
|
Lu T, Hu X, van Haren MHI, Spruijt E, Huck WTS. Structure-Property Relationships Governing Membrane-Penetrating Behaviour of Complex Coacervates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303138. [PMID: 37218010 DOI: 10.1002/smll.202303138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Indexed: 05/24/2023]
Abstract
Complex coacervates are phase-separated liquid droplets composed of oppositely charged multivalent molecules. The unique material properties of the complex coacervate interior favours the sequestration of biomolecules and facilitates reactions. Recently, it is shown that coacervates can be used for direct cytosolic delivery of sequestered biomolecules in living cells. Here, it is studied that the physical properties required for complex coacervates composed of oligo-arginine and RNA to cross phospholipid bilayers and enter liposomes penetration depends on two main parameters: the difference in ζ-potential between the complex coacervates and the liposomes, and the partitioning coefficient (Kp ) of lipids into the complex coacervates. Following these guidelines, a range of complex coacervates is found that is able to penetrate the membrane of living cells, thus paving the way for further development of coacervates as delivery vehicles of therapeutic agents.
Collapse
Affiliation(s)
- Tiemei Lu
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, the Netherlands
| | - Xinyu Hu
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, the Netherlands
| | - Merlijn H I van Haren
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, the Netherlands
| | - Evan Spruijt
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, the Netherlands
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, 6525 AJ, the Netherlands
| |
Collapse
|
22
|
Ma H, Xing F, Zhou Y, Yu P, Luo R, Xu J, Xiang Z, Rommens PM, Duan X, Ritz U. Design and fabrication of intracellular therapeutic cargo delivery systems based on nanomaterials: current status and future perspectives. J Mater Chem B 2023; 11:7873-7912. [PMID: 37551112 DOI: 10.1039/d3tb01008b] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Intracellular cargo delivery, the introduction of small molecules, proteins, and nucleic acids into a specific targeted site in a biological system, is an important strategy for deciphering cell function, directing cell fate, and reprogramming cell behavior. With the advancement of nanotechnology, many researchers use nanoparticles (NPs) to break through biological barriers to achieving efficient targeted delivery in biological systems, bringing a new way to realize efficient targeted drug delivery in biological systems. With a similar size to many biomolecules, NPs possess excellent physical and chemical properties and a certain targeting ability after functional modification on the surface of NPs. Currently, intracellular cargo delivery based on NPs has emerged as an important strategy for genome editing regimens and cell therapy. Although researchers can successfully deliver NPs into biological systems, many of them are delivered very inefficiently and are not specifically targeted. Hence, the development of efficient, target-capable, and safe nanoscale drug delivery systems to deliver therapeutic substances to cells or organs is a major challenge today. In this review, on the basis of describing the research overview and classification of NPs, we focused on the current research status of intracellular cargo delivery based on NPs in biological systems, and discuss the current problems and challenges in the delivery process of NPs in biological systems.
Collapse
Affiliation(s)
- Hong Ma
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Fei Xing
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Ludwigstraße 23, 35392 Giessen, Germany
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Rong Luo
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiawei Xu
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Xin Duan
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedic Surgery, The Fifth People's Hospital of Sichuan Province, Chengdu, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
23
|
Tomazini A, Shifman JM. Targeting Ras with protein engineering. Oncotarget 2023; 14:672-687. [PMID: 37395750 DOI: 10.18632/oncotarget.28469] [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: 07/04/2023] Open
Abstract
Ras proteins are small GTPases that regulate cell growth and division. Mutations in Ras genes are associated with many types of cancer, making them attractive targets for cancer therapy. Despite extensive efforts, targeting Ras proteins with small molecules has been extremely challenging due to Ras's mostly flat surface and lack of small molecule-binding cavities. These challenges were recently overcome by the development of the first covalent small-molecule anti-Ras drug, sotorasib, highlighting the efficacy of Ras inhibition as a therapeutic strategy. However, this drug exclusively inhibits the Ras G12C mutant, which is not a prevalent mutation in most cancer types. Unlike the G12C variant, other Ras oncogenic mutants lack reactive cysteines, rendering them unsuitable for targeting via the same strategy. Protein engineering has emerged as a promising method to target Ras, as engineered proteins have the ability to recognize various surfaces with high affinity and specificity. Over the past few years, scientists have engineered antibodies, natural Ras effectors, and novel binding domains to bind to Ras and counteract its carcinogenic activities via a variety of strategies. These include inhibiting Ras-effector interactions, disrupting Ras dimerization, interrupting Ras nucleotide exchange, stimulating Ras interaction with tumor suppressor genes, and promoting Ras degradation. In parallel, significant advancements have been made in intracellular protein delivery, enabling the delivery of the engineered anti-Ras agents into the cellular cytoplasm. These advances offer a promising path for targeting Ras proteins and other challenging drug targets, opening up new opportunities for drug discovery and development.
Collapse
Affiliation(s)
- Atilio Tomazini
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Julia M Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
24
|
Deng F, Li Y, Hall T, Vesey G, Goldys EM. Bi-functional antibody-CRISPR/Cas12a ribonucleoprotein conjugate for improved immunoassay performance. Anal Chim Acta 2023; 1259:341211. [PMID: 37100476 DOI: 10.1016/j.aca.2023.341211] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/27/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023]
Abstract
Protein conjugates are commonly used in biochemistry, including diagnostic platforms such as antibody-based immunoassays. Antibodies can be bound to a variety of molecules creating conjugates with desirable functions, particularly for imaging and signal amplification. Cas12a is a recently discovered programable nuclease with the remarkable capability to amplify assay signals due to its trans-cleavage property. In this study, we directly conjugated antibody with Cas12a/gRNA ribonucleoprotein without the loss of function in either constituent. The conjugated antibody was suitable for immunoassays and the conjugated Cas12a was capable of amplifying the signal produced in an immunosensor without the need to change the original assay protocol. We applied the bi-functional antibody-Cas12a/gRNA conjugate to successfully detect two different types of targets, a whole pathogenic microorganism, Cryptosporidium, and a small protein, cytokine IFN-γ, with sensitivity reaching one single microorganism per sample and 10 fg/mL for IFN-γ, respectively. With simple substitution of the antibody conjugated with the Cas12a/gRNA RNP, this approach can potentially be applied to increase sensitivity of a variety of immunoassays for a broad range of different analytes.
Collapse
Affiliation(s)
- Fei Deng
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia; ARC Centre of Excellence in Nanoscale Biophotonics, University of New South Wales, Sydney, 2052, Australia.
| | - Yi Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia; ARC Centre of Excellence in Nanoscale Biophotonics, University of New South Wales, Sydney, 2052, Australia.
| | - Tim Hall
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Graham Vesey
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia
| | - Ewa M Goldys
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2052, Australia; ARC Centre of Excellence in Nanoscale Biophotonics, University of New South Wales, Sydney, 2052, Australia
| |
Collapse
|
25
|
Porello I, Cellesi F. Intracellular delivery of therapeutic proteins. New advancements and future directions. Front Bioeng Biotechnol 2023; 11:1211798. [PMID: 37304137 PMCID: PMC10247999 DOI: 10.3389/fbioe.2023.1211798] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Achieving the full potential of therapeutic proteins to access and target intracellular receptors will have enormous benefits in advancing human health and fighting disease. Existing strategies for intracellular protein delivery, such as chemical modification and nanocarrier-based protein delivery approaches, have shown promise but with limited efficiency and safety concerns. The development of more effective and versatile delivery tools is crucial for the safe and effective use of protein drugs. Nanosystems that can trigger endocytosis and endosomal disruption, or directly deliver proteins into the cytosol, are essential for successful therapeutic effects. This article aims to provide a brief overview of the current methods for intracellular protein delivery to mammalian cells, highlighting current challenges, new developments, and future research opportunities.
Collapse
|
26
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
27
|
Wang B, Zhang P, Wang Q, Zou S, Song J, Zhang F, Liu G, Zhang L. Protective Effects of a Jellyfish-Derived Thioredoxin Fused with Cell-Penetrating Peptide TAT-PTD on H 2O 2-Induced Oxidative Damage. Int J Mol Sci 2023; 24:ijms24087340. [PMID: 37108504 PMCID: PMC10138494 DOI: 10.3390/ijms24087340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Thioredoxin (Trx) plays a critical role in maintaining redox balance in various cells and exhibits anti-oxidative, anti-apoptotic, and anti-inflammatory effects. However, whether exogenous Trx can inhibit intracellular oxidative damage has not been investigated. In previous study, we have identified a novel Trx from the jellyfish Cyanea capillata, named CcTrx1, and confirmed its antioxidant activities in vitro. Here, we obtained a recombinant protein, PTD-CcTrx1, which is a fusion of CcTrx1 and protein transduction domain (PTD) of HIV TAT protein. The transmembrane ability and antioxidant activities of PTD-CcTrx1, and its protective effects against H2O2-induced oxidative damage in HaCaT cells were also detected. Our results revealed that PTD-CcTrx1 exhibited specific transmembrane ability and antioxidant activities, and it could significantly attenuate the intracellular oxidative stress, inhibit H2O2-induced apoptosis, and protect HaCaT cells from oxidative damage. The present study provides critical evidence for application of PTD-CcTrx1 as a novel antioxidant to treat skin oxidative damage in the future.
Collapse
Affiliation(s)
- Bo Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Center, Naval Medical University, Shanghai 200433, China
- Department of Infectious Disease, No. 971 Hospital of the PLA Navy, Qingdao 266071, China
| | - Peipei Zhang
- Department of Marine Biological Injury and Dermatology, Naval Special Medical Center, Naval Medical University, Shanghai 200052, China
| | - Qianqian Wang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Shuaijun Zou
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Juxingsi Song
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Fuhai Zhang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Guoyan Liu
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Liming Zhang
- Department of Marine Biomedicine and Polar Medicine, Naval Special Medical Center, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
28
|
Son H, Shin J, Park J. Recent progress in nanomedicine-mediated cytosolic delivery. RSC Adv 2023; 13:9788-9799. [PMID: 36998521 PMCID: PMC10043881 DOI: 10.1039/d2ra07111h] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Cytosolic delivery of bioactive agents has exhibited great potential to cure undruggable targets and diseases. Because biological cell membranes are a natural barrier for living cells, efficient delivery methods are required to transfer bioactive and therapeutic agents into the cytosol. Various strategies that do not require cell invasive and harmful processes, such as endosomal escape, cell-penetrating peptides, stimuli-sensitive delivery, and fusogenic liposomes, have been developed for cytosolic delivery. Nanoparticles can easily display functionalization ligands on their surfaces, enabling many bio-applications for cytosolic delivery of various cargo, including genes, proteins, and small-molecule drugs. Cytosolic delivery uses nanoparticle-based delivery systems to avoid degradation of proteins and keep the functionality of other bioactive molecules, and functionalization of nanoparticle-based delivery vehicles imparts a specific targeting ability. With these advantages, nanomedicines have been used for organelle-specific tagging, vaccine delivery for enhanced immunotherapy, and intracellular delivery of proteins and genes. Optimization of the size, surface charges, specific targeting ability, and composition of nanoparticles is needed for various cargos and target cells. Toxicity issues with the nanoparticle material must be managed to enable clinical use.
Collapse
Affiliation(s)
- Hangyu Son
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea 222 Banpo-daero, Seocho-gu Seoul 06591 Republic of Korea
| | - Jeongsu Shin
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea 222 Banpo-daero, Seocho-gu Seoul 06591 Republic of Korea
| | - Joonhyuck Park
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea 222 Banpo-daero, Seocho-gu Seoul 06591 Republic of Korea
| |
Collapse
|
29
|
Wang C, Xiao C, Chen Y, Li Y, Zhang Q, Shan W, Li Y, Bi S, Wang Y, Wang X, Ren L. Sequential administration of virus-like particle-based nanomedicine to elicit enhanced tumor chemotherapy. J Mater Chem B 2023; 11:2674-2683. [PMID: 36857702 DOI: 10.1039/d2tb02163c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Protein cages have played a long-standing role in biomedicine applications, especially in tumor chemotherapy. Among protein cages, virus like particles (VLPs) have received attention for their potential applications in vaccine development and targeted drug delivery. However, most of the existing protein-based platform technologies are plagued with immunological problems that may limit their systemic delivery efficiency as drug carriers. Here, we show that using immune-orthogonal protein cages sequentially and modifying the dominant loop epitope can circumvent adaptive immune responses and enable effective drug delivery using repeated dosing. We genetically modified three different hepadnavirus core protein derived VLPs as delivery vectors for doxorubicin (DOX). These engineered VLPs have similar assembly characteristics, particle sizes, and immunological properties. Our results indicated that there was negligible antibody cross-reactivity in either direction between these three RGD-VLPs in mice that were previously immunized against HBc VLPs. Moreover, the sequential administration of multiple RGD-VLP-based nanomedicine (DOX@RGD-VLPs) could effectively reduce immune clearance and inhibited tumor growth. Hence, this study could provide an attractive protein cage-based platform for therapeutic drug delivery.
Collapse
Affiliation(s)
- Chufan Wang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Cheng Xiao
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Yurong Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Yao Li
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Qiang Zhang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
| | - Wenjun Shan
- Department of Pharmacology, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing 400038, P. R. China
| | - Yulin Li
- Henan Bioengineering Research Center, Zhengdong New District, Zhengzhou, China
| | - Shengli Bi
- Chinese Center for Disease Control & Prevention, Institute Viral Disease Control & Prevention, Beijing, P. R. China
| | - Yunlong Wang
- Henan Bioengineering Research Center, Zhengdong New District, Zhengzhou, China
| | - Xiumin Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Lei Ren
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, P. R. China
- State Key Lab of Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, P. R. China
| |
Collapse
|
30
|
Um H, Kang RH, Kim D. Iron-silicate-coated porous silicon nanoparticles for in situ ROS self-generation. Colloids Surf B Biointerfaces 2023; 225:113273. [PMID: 36965332 DOI: 10.1016/j.colsurfb.2023.113273] [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: 12/19/2022] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023]
Abstract
Porous silicon nanoparticles (pSiNPs) have gained attention from drug delivery systems (DDS) due to their biocompatibility, high drug-loading efficiency, and facile surface modification. To date, many surface chemistries of pSiNPs have been developed to maximize the merits and overcome the drawbacks of pSiNPs. In this work, we newly disclosed a formulation, iron-silicate-coated pSiNPs (Fe-pSiNPs-NCS), using the surface modification method with iron-silicate and 3-isothiocyanatopropyltriethoxysilane (TEPITC). Fe-pSiNPs-NCS demonstrated effective reactive-oxygen species (ROS) self-generation ability via a Fenton-like reaction of iron-silicate and in situ hydrogen peroxide (H2O2) generation of TEPITC on the surface of pSiNPs, resulting in excellent anticancer effect in U87MG cancer cells. Moreover, we confirmed that Fe-pSiNPs-NCS could be used as a drug delivery carrier as it was proven that anticancer drugs (doxorubicin, SN-38) were loaded into Fe-pSiNPs-NCS with high-loading efficiency. These findings could offer efficient strategies for developing nanotherapeutics in biomedical fields.
Collapse
Affiliation(s)
- Hyeji Um
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, the Republic of Korea
| | - Rae Hyung Kang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, the Republic of Korea; Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University, Seoul 02447, the Republic of Korea; Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, the Republic of Korea; Center for Converging Humanities, Kyung Hee University, Seoul 02447, the Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, the Republic of Korea; UC San Diego Materials Research Science and Engineering Center, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
31
|
Lv J, Yang Z, Wang C, Duan J, Ren L, Rong G, Feng Q, Li Y, Cheng Y. Efficient intracellular and in vivo delivery of toxin proteins by a ROS-responsive polymer for cancer therapy. J Control Release 2023; 355:160-170. [PMID: 36736906 DOI: 10.1016/j.jconrel.2023.01.064] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/17/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Rational design of efficient cytosolic protein delivery carriers holds enormous promise for biotherapeutics development. Several delivery systems have been developed during the past decades, while tailoring the balance between extracellular protein binding and intracellular cargo release is still challenging. In this study, we synthesized a series of oxygen-sensitive reactive polymers, rich in boron, by radical polymerization and post-modification for cytosolic protein delivery in vitro and in vivo. The introduction of boronate building blocks into the polymer scaffold significantly enhanced its protein binding affinity, and the polymer/protein complexes with high stability were obtained by tailoring the molecular ratios between the boronate ligands and the amine groups. The lead material screened from the polymer library exhibited efficient protein delivery efficacy that can release cargo proteins in cytosol in a reactive oxygen species responsive manner, which enables intracellular delivery of proteins with maintained bioactivity. In addition, the polymer-based nanoformulations efficiently delivered saporin, a toxin protein, into osteosarcoma cells and tumor tissues, and exhibited high therapeutic efficacy in an osteosarcoma mouse model. The synthesized polymer in this study can be developed as a promising nanocarrier for cytosolic delivery of protein therapeutics to treat a variety of diseases.
Collapse
Affiliation(s)
- Jia Lv
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhen Yang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Changping Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jianan Duan
- South China Advanced Institute for Soft Matter Science and Technology, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
| | - Lanfang Ren
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Guangyu Rong
- South China Advanced Institute for Soft Matter Science and Technology, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
| | - Qiuyu Feng
- South China Advanced Institute for Soft Matter Science and Technology, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, School of Emergent Soft Matter, South China University of Technology, Guangzhou 510640, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
32
|
Chen C, Gao P, Wang H, Cheng Y, Lv J. Histidine-based coordinative polymers for efficient intracellular protein delivery via enhanced protein binding, cellular uptake, and endosomal escape. Biomater Sci 2023; 11:1765-1775. [PMID: 36648450 DOI: 10.1039/d2bm01541b] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polymers are one of the most promising protein delivery carriers; however, their applications are hindered by low delivery efficacy owing to their undesirable performance in protein binding, cellular uptake and endosomal escape. Here, we designed a series of histidine-based coordinative polymers for efficient intracellular protein delivery. Coordination of metal ions such as Ni2+, Zn2+, and Cu2+ with histidine residues on a polymer greatly improved its performance in protein binding, complex stability, cellular uptake and endosomal escape, therefore achieving highly improved protein delivery efficacy. Among the coordinative polymers, the Zn2+-coordinated one exhibited the highest cellular uptake, while the Cu2+-coordinated one exhibited the highest endosomal escape. The Ni2+-coordinated polymer formed large-sized aggregates with cargo proteins and showed insufficient protein release after endocytosis. The results obtained in this study provided new insight into the development of coordinative polymer-based protein delivery systems.
Collapse
Affiliation(s)
- Changyuan Chen
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Peng Gao
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Jia Lv
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
33
|
Lam KK, Wong SH, Cheah PY. Targeting the 'Undruggable' Driver Protein, KRAS, in Epithelial Cancers: Current Perspective. Cells 2023; 12:cells12040631. [PMID: 36831298 PMCID: PMC9954350 DOI: 10.3390/cells12040631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/30/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
This review summarizes recent development in synthetic drugs and biologics targeting intracellular driver genes in epithelial cancers, focusing on KRAS, and provides a current perspective and potential leads for the field. Compared to biologics, small molecule inhibitors (SMIs) readily penetrate cells, thus being able to target intracellular proteins. However, SMIs frequently suffer from pleiotropic effects, off-target cytotoxicity and invariably elicit resistance. In contrast, biologics are much larger molecules limited by cellular entry, but if this is surmounted, they may have more specific effects and less therapy-induced resistance. Exciting breakthroughs in the past two years include engineering of non-covalent KRAS G12D-specific inhibitor, probody bispecific antibodies, drug-peptide conjugate as MHC-restricted neoantigen to prompt immune response by T-cells, and success in the adoptive cell therapy front in both breast and pancreatic cancers.
Collapse
Affiliation(s)
- Kuen Kuen Lam
- Department of Colorectal Surgery, Singapore General Hospital, Singapore 169856, Singapore
| | | | - Peh Yean Cheah
- Department of Colorectal Surgery, Singapore General Hospital, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
- Correspondence:
| |
Collapse
|
34
|
Goswami R, Lehot V, Çiçek YA, Nagaraj H, Jeon T, Nguyen T, Fedeli S, Rotello VM. Direct Cytosolic Delivery of Citraconylated Proteins. Pharmaceutics 2023; 15:pharmaceutics15010218. [PMID: 36678847 PMCID: PMC9861219 DOI: 10.3390/pharmaceutics15010218] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/11/2023] Open
Abstract
Current intracellular protein delivery strategies face the challenge of endosomal entrapment and consequent degradation of protein cargo. Methods to efficiently deliver proteins directly to the cytosol have the potential to overcome this hurdle. Here, we report the use of a straightforward approach of protein modification using citraconic anhydride to impart an overall negative charge on the proteins, enabling them to assemble with positively charged nano vectors. This strategy uses anhydride-modified proteins to electrostatically form polymer-protein nanocomposites with a cationic guanidinium-functionalized polymer. These supramolecular self-assemblies demonstrated the efficient cytosolic delivery of modified proteins through a membrane fusion-like mechanism. This approach was validated on five cell lines and seven proteins as cargo. Retention of protein function was confirmed through efficient cell killing via the intracellular enzymatic activity of RNase A. This platform provides a versatile, straightforward, and single-step method of protein modification and efficient direct cytosolic protein delivery.
Collapse
Affiliation(s)
- Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Victor Lehot
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Yağız Anıl Çiçek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Terry Nguyen
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
- Correspondence:
| |
Collapse
|
35
|
Shrestha B, Tang L, Hood RL. Nanotechnology for Personalized Medicine. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
36
|
Kapelner RA, Fisher RS, Elbaum-Garfinkle S, Obermeyer AC. Protein charge parameters that influence stability and cellular internalization of polyelectrolyte complex micelles. Chem Sci 2022; 13:14346-14356. [PMID: 36545145 PMCID: PMC9749388 DOI: 10.1039/d2sc00192f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Proteins are an important class of biologics, but there are several recurring challenges to address when designing protein-based therapeutics. These challenges include: the propensity of proteins to aggregate during formulation, relatively low loading in traditional hydrophobic delivery vehicles, and inefficient cellular uptake. This last criterion is particularly challenging for anionic proteins as they cannot cross the anionic plasma membrane. Here we investigated the complex coacervation of anionic proteins with a block copolymer of opposite charge to form polyelectrolyte complex (PEC) micelles for use as a protein delivery vehicle. Using genetically modified variants of the model protein green fluorescent protein (GFP), we evaluated the role of protein charge and charge localization in the formation and stability of PEC micelles. A neutral-cationic block copolymer, poly(oligoethylene glycol methacrylate-block-quaternized 4-vinylpyridine), POEGMA79-b-qP4VP175, was prepared via RAFT polymerization for complexation and microphase separation with the panel of engineered anionic GFPs. We found that isotropically supercharged proteins formed micelles at higher ionic strength relative to protein variants with charge localized to a polypeptide tag. We then studied GFP delivery by PEC micelles and found that they effectively delivered the protein cargo to mammalian cells. However, cellular delivery varied as a function of protein charge and charge distribution and we found an inverse relationship between the PEC micelle critical salt concentration and delivery efficiency. This model system has highlighted the potential of polyelectrolyte complexes to deliver anionic proteins intracellularly. Using this model system, we have identified requirements for the formation of PEC micelles that are stable at physiological ionic strength and that smaller protein-polyelectrolyte complexes effectively deliver proteins to Jurkat cells.
Collapse
Affiliation(s)
- Rachel A Kapelner
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
| | - Rachel S Fisher
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
- Structural Biology Initiative, CUNY Advanced Science Research Center New York NY USA
| | - Shana Elbaum-Garfinkle
- Structural Biology Initiative, CUNY Advanced Science Research Center New York NY USA
- PhD Programs in Biochemistry and Biology at the Graduate Center, City University of New York NY USA
| | - Allie C Obermeyer
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
| |
Collapse
|
37
|
Li X, Ren X, Zhang Y, Ding L, Huo M, Li Q. Fabry disease: Mechanism and therapeutics strategies. Front Pharmacol 2022; 13:1025740. [PMID: 36386210 PMCID: PMC9643830 DOI: 10.3389/fphar.2022.1025740] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Fabry disease is a monogenic disease characterized by a deficiency or loss of the α-galactosidase A (GLA). The resulting impairment in lysosomal GLA enzymatic activity leads to the pathogenic accumulation of enzymatic substrate and, consequently, the progressive appearance of clinical symptoms in target organs, including the heart, kidney, and brain. However, the mechanisms involved in Fabry disease-mediated organ damage are largely ambiguous and poorly understood, which hinders the development of therapeutic strategies for the treatment of this disorder. Although currently available clinical approaches have shown some efficiency in the treatment of Fabry disease, they all exhibit limitations that need to be overcome. In this review, we first introduce current mechanistic knowledge of Fabry disease and discuss potential therapeutic strategies for its treatment. We then systemically summarize and discuss advances in research on therapeutic approaches, including enzyme replacement therapy (ERT), gene therapy, and chaperone therapy, as well as strategies targeting subcellular compartments, such as lysosomes, the endoplasmic reticulum, and the nucleus. Finally, the future development of potential therapeutic strategies is discussed based on the results of mechanistic studies and the limitations associated with these therapeutic approaches.
Collapse
Affiliation(s)
- Xi Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiangyi Ren
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Yabing Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Lin Ding
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Minfeng Huo
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| |
Collapse
|
38
|
Cheng Z, Li Y, Zhao D, Zhao W, Wu M, Zhang W, Cui Y, Zhang P, Zhang Z. Nanocarriers for intracellular co-delivery of proteins and small-molecule drugs for cancer therapy. Front Bioeng Biotechnol 2022; 10:994655. [PMID: 36147526 PMCID: PMC9485877 DOI: 10.3389/fbioe.2022.994655] [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: 07/15/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In the past few decades, the combination of proteins and small-molecule drugs has made tremendous progress in cancer treatment, but it is still not satisfactory. Because there are great differences in molecular weight, water solubility, stability, pharmacokinetics, biodistribution, and the ways of release and action between macromolecular proteins and small-molecule drugs. To improve the efficacy and safety of tumor treatment, people are committed to developing protein and drug co-delivery systems. Currently, intracellular co-delivery systems have been developed that integrate proteins and small-molecule drugs into one nanocarrier via various loading strategies. These systems significantly improve the blood stability, half-life, and biodistribution of proteins and small-molecule drugs, thus increasing their concentration in tumors. Furthermore, proteins and small-molecule drugs within these systems can be specifically targeted to tumor cells, and are released to perform functions after entering tumor cells simultaneously, resulting in improved effectiveness and safety of tumor treatment. This review summarizes the latest progress in protein and small-molecule drug intracellular co-delivery systems, with emphasis on the composition of nanocarriers, as well as on the loading methods of proteins and small-molecule drugs that play a role in cells into the systems, which have not been summarized by others so far.
Collapse
Affiliation(s)
- Zhihong Cheng
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yongshuang Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Meng Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Weilin Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
39
|
Chen P, Yang W, Hong T, Miyazaki T, Dirisala A, Kataoka K, Cabral H. Nanocarriers escaping from hyperacidified endo/lysosomes in cancer cells allow tumor-targeted intracellular delivery of antibodies to therapeutically inhibit c-MYC. Biomaterials 2022; 288:121748. [PMID: 36038419 DOI: 10.1016/j.biomaterials.2022.121748] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/02/2022]
Abstract
Intracellular protein delivery is a powerful strategy for developing innovative therapeutics. Nanocarriers present great potential to deliver proteins inside cells by promoting cellular uptake and overcoming entrapment and degradation in acidic endo/lysosomal compartments. Thus, because cytosolic access is essential for eliciting the function of proteins, significant efforts have been dedicated to engineering nanocarriers with maximal endosomal escape regardless of the cell type. On the other hand, controlling the ability of nanocarriers to escape from the endo/lysosomal compartments of particular cells may offer the opportunity for enhancing delivery precision. To test this hypothesis, we developed pH-sensitive polymeric nanocarriers with adjustable endosomal escape potency for selectively reaching the cytosol of defined cancer cells with dysregulated endo/lysosomal acidification. By loading antibodies against nuclear pore complex in the nanocarriers, we demonstrated the selective delivery into the cytosol and subsequent nucleus targeting of cancer cells rather than non-cancerous cells both in vitro and in vivo. Systemically injected nanocarriers loading anti-c-MYC antibodies suppressed c-MYC in solid tumors and inhibit tumor growth without side effects, confirming the therapeutic potential of our approach. These results indicated that regulating the ability of nanocarriers to escape from endo/lysosomal compartments in particular cells is a practical approach for gaining delivery specificity.
Collapse
Affiliation(s)
- Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Wenqian Yang
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taehun Hong
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takuya Miyazaki
- Kanagawa Institute of Industrial Science and Technology, 705-1 Shimoimaizumi, Ebina City, Kanagawa, 243-0435, Japan
| | - Anjaneyulu Dirisala
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
40
|
Xiong J, Chu JCH, Fong WP, Wong CTT, Ng DKP. Specific Activation of Photosensitizer with Extrinsic Enzyme for Precisive Photodynamic Therapy. J Am Chem Soc 2022; 144:10647-10658. [PMID: 35639988 DOI: 10.1021/jacs.2c04017] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Delivery of functional proteins into the intracellular space has been a challenging task that could lead to a myriad of therapeutic applications. We report herein a novel bioconjugation strategy for enzyme modification and selective delivery into cancer cells for lock-and-key-type activation of photosensitizers. Using a bifunctional linker containing a bis(bromomethyl)phenyl group and an o-phthalaldehyde moiety, it could induce cyclization of the peptide sequence Ac-NH-CRGDfC-CONH2 through site-specific dibenzylation with the two cysteine residues and further coupling with β-galactosidase via the phthalaldehyde-amine capture reaction. This facile two-step one-pot procedure enabled the preparation of cyclic RGD-modified β-galactosidase readily, which could be internalized selectively into αvβ3 integrin-overexpressed cancer cells. Upon encountering an intrinsically quenched distyryl boron dipyrromethene-based photosensitizer conjugated with a galactose moiety through a self-immolative linker inside the cells, the extrinsic enzyme induced specific cleavage of the β-galactosidic bond followed by self-immolation to release an activated derivative, thereby restoring the photodynamic activities and causing cell death effectively. The high specificity of this extrinsic enzyme-activated photosensitizing system was also demonstrated in vivo using nude mice bearing an αvβ3 integrin-positive U87-MG tumor. The specific activation at the tumor site resulted in lighting up and complete eradication of the tumor upon laser irradiation, while by using the native β-galactosidase, the effects were largely reduced. In contrast to the conventional activation using intrinsic enzymes, this extrinsic enzyme activatable approach can further minimize the nonspecific activation toward precisive photodynamic therapy.
Collapse
Affiliation(s)
- Junlong Xiong
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Jacky C H Chu
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Wing-Ping Fong
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Clarence T T Wong
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Dennis K P Ng
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| |
Collapse
|
41
|
Chen Z, Zhang J, Lyu Q, Wang H, Ji X, Yan Z, Chen F, Dahlgren RA, Zhang M. Modular configurations of living biomaterials incorporating nano-based artificial mediators and synthetic biology to improve bioelectrocatalytic performance: A review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 824:153857. [PMID: 35176368 DOI: 10.1016/j.scitotenv.2022.153857] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/24/2022] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
Currently, the industrial application of bioelectrochemical systems (BESs) that are incubated with natural electrochemically active microbes (EABs) is limited due to inefficient extracellular electron transfer (EET) by natural EABs. Notably, recent studies have identified several novel living biomaterials comprising highly efficient electron transfer systems allowing unparalleled proficiency of energy conversion. Introduction of these biomaterials into BESs could fundamentally increase their utilization for a wide range of applications. This review provides a comprehensive assessment of recent advancements in the design of living biomaterials that can be exploited to enhance bioelectrocatalytic performance. Further, modular configurations of abiotic and biotic components promise a powerful enhancement through integration of nano-based artificial mediators and synthetic biology. Herein, recent advancements in BESs are synthesized and assessed, including heterojunctions between conductive nanomaterials and EABs, in-situ hybrid self-assembly of EABs and nano-sized semiconductors, cytoprotection in biohybrids, synthetic biological modifications of EABs and electroactive biofilms. Since living biomaterials comprise a broad range of disciplines, such as molecular biology, electrochemistry and material sciences, full integration of technological advances applied in an interdisciplinary framework will greatly enhance/advance the utility and novelty of BESs. Overall, emerging fundamental knowledge concerning living biomaterials provides a powerful opportunity to markedly boost EET efficiency and facilitate the industrial application of BESs to meet global sustainability challenges/goals.
Collapse
Affiliation(s)
- Zheng Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, People's Republic of China; School of Environmental Science & Engineering, Tan Kah Kee College, Xiamen University, Zhangzhou 363105, People's Republic of China; Fujian Provincial Key Lab of Coastal Basin Environment, Fujian Polytechnic Normal University, Fuqing 350300, People's Republic of China.
| | - Jing Zhang
- School of Environmental Science & Engineering, Tan Kah Kee College, Xiamen University, Zhangzhou 363105, People's Republic of China
| | - Qingyang Lyu
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, People's Republic of China
| | - Honghui Wang
- School of Environmental Science & Engineering, Tan Kah Kee College, Xiamen University, Zhangzhou 363105, People's Republic of China
| | - Xiaoliang Ji
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, People's Republic of China
| | - Zhiying Yan
- CAS Key Laboratory of Environmental and Applied Microbiology, Environmental Microbiology Key Laboratory of Sichuan Province, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, People's Republic of China
| | - Fang Chen
- Fujian Provincial Key Lab of Coastal Basin Environment, Fujian Polytechnic Normal University, Fuqing 350300, People's Republic of China
| | - Randy A Dahlgren
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, People's Republic of China; Department of Land, Air and Water Resources, University of California, Davis, CA 95616, USA
| | - Minghua Zhang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, People's Republic of China; Department of Land, Air and Water Resources, University of California, Davis, CA 95616, USA
| |
Collapse
|
42
|
Luther DC, Nagaraj H, Goswami R, Çiçek YA, Jeon T, Gopalakrishnan S, Rotello VM. Direct Cytosolic Delivery of Proteins Using Lyophilized and Reconstituted Polymer-Protein Assemblies. Pharm Res 2022; 39:1197-1204. [PMID: 35297498 PMCID: PMC10587898 DOI: 10.1007/s11095-022-03226-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: 11/24/2021] [Accepted: 03/04/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Cytosolic delivery of proteins accesses intracellular targets for chemotherapy and immunomodulation. Current delivery systems utilize inefficient endosomal pathways of uptake and escape that lead to degradation of delivered cargo. Cationic poly(oxanorbornene)imide (PONI) polymers enable highly efficient cytosolic delivery of co-engineered proteins, but aggregation and denaturation in solution limits shelf life. In the present study we evaluate polymer-protein nanocomposite vehicles as candidates for lyophilization and point-of-care resuspension to provide a transferrable technology for cytosolic protein delivery. METHODS Self-assembled nanocomposites of engineered poly(glutamate)-tagged (E-tagged) proteins and guanidinium-functionalized PONI homopolymers were generated, lyophilized, and stored for 2 weeks. After reconstitution and delivery, cytosolic access of E-tagged GFP cargo (GFPE15) was assessed through diffuse cytosolic and nuclear fluorescence, and cell killing with chemotherapeutic enzyme Granzyme A (GrAE10). Efficiency was quantified between freshly prepared and lyophilized samples. RESULTS Reconstituted nanocomposites retained key structural features of freshly prepared assemblies, with minimal loss of material. Cytosolic delivery (> 80% efficiency of freshly prepared nanocomposites) of GFPE15 was validated in several cell lines, with intracellular access validated and quantified through diffusion into the nucleus. Delivery of GrAE10 elicited significant tumorigenic cell death. Intracellular access of cytotoxic protein was validated through cell viability. CONCLUSION Reconstituted nanocomposites achieved efficient cytosolic delivery of protein cargo and demonstrated therapeutic applicability with delivery of GrAE10. Overall, this strategy represents a versatile and highly translatable method for cytosolic delivery of proteins.
Collapse
Affiliation(s)
- David C Luther
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Yağız Anıl Çiçek
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts, 379A LGRT Tower A, 710 North Pleasant St., Massachusetts, 01003, Amherst, USA.
| |
Collapse
|
43
|
Luther DC, Lee YW, Nagaraj H, Clark V, Jeon T, Goswami R, Gopalakrishnan S, Fedeli S, Jerome W, Elia JL, Rotello VM. Cytosolic Protein Delivery Using Modular Biotin-Streptavidin Assembly of Nanocomposites. ACS NANO 2022; 16:7323-7330. [PMID: 35435664 PMCID: PMC10586328 DOI: 10.1021/acsnano.1c06768] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Current strategies for the delivery of proteins into cells face general challenges of endosomal entrapment and concomitant degradation of protein cargo. Efficient delivery directly to the cytosol overcomes this obstacle: we report here the use of biotin-streptavidin tethering to provide a modular approach to the generation of nanovectors capable of a cytosolic delivery of biotinylated proteins. This strategy uses streptavidin to organize biotinylated protein and biotinylated oligo(glutamate) peptide into modular complexes that are then electrostatically self-assembled with a cationic guanidinium-functionalized polymer. The resulting polymer-protein nanocomposites demonstrate efficient cytosolic delivery of six biotinylated protein cargos of varying size, charge, and quaternary structure. Retention of protein function was established through efficient cell killing via delivery of the chemotherapeutic enzyme granzyme A. This platform represents a versatile and modular approach to intracellular delivery through the noncovalent tethering of multiple components into a single delivery vector.
Collapse
Affiliation(s)
- David C. Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent Clark
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Sanjana Gopalakrishnan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - William Jerome
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - James L. Elia
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
44
|
De R, Song YH, Mahata MK, Lee KT. pH-responsive polyelectrolyte complexation on upconversion nanoparticles: a multifunctional nanocarrier for protection, delivery, and 3D-imaging of therapeutic protein. J Mater Chem B 2022; 10:3420-3433. [PMID: 35389393 DOI: 10.1039/d2tb00246a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The delicate tertiary structure of proteins, their susceptibility to heat- and enzyme-induced irreversible denaturation, and their tendency to get accumulated at the cell membrane during uptake are daunting challenges in proteinaceous therapeutic delivery. Herein, a polyelectrolyte complex having encapsulated therapeutic protein has been designed on the surface of upconverting luminescent nanoparticles (NaYF4:20%Yb3+,2%Er3+). This nanosized complex system has been found to overcome the challenges of protein aggregation at the cell membrane. It has also defended the cargo from denaturation against (a) enzymatic action of proteinase K and (b) heat (up to 60 °C). Additionally, the nanoparticles at the core of the loaded carrier served as near-infrared (980 nm) responsive probe to accomplish extended-duration 3D imaging during protein delivery. The outer layer of polymer played pivotal role to protect/retrieve the protein structure from denaturation as investigated by circular dichroism studies. Both the masked surface-charges of protein and the nanoscale size of the loaded carrier have facilitated their efficient passage through the cell membrane as observed through 3D images/videos. This nanocarrier is the first of its kind for direct delivery of protein. Thus, the findings can be useful to protect and transport various proteinaceous materials to overcome challenges of accumulation at the cell-membrane and low-temperature storage, as nature does.
Collapse
Affiliation(s)
- Ranjit De
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, South Korea. .,Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Yo Han Song
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, South Korea.
| | - Manoj Kumar Mahata
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, South Korea. .,Drittes Physikalisches Institut - Biophysik, Georg-August-Universität Göttingen, Friedrich-Hund-Platz 1, 37077, Göttingen, Germany
| | - Kang Taek Lee
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, South Korea.
| |
Collapse
|
45
|
Song Y, Dai B, Wang Y, Wang Y, Liu C, Gourdon P, Liu L, Wang K, Dong M. Identifying Heterozipper β-Sheet in Twisted Amyloid Aggregation. NANO LETTERS 2022; 22:3707-3712. [PMID: 35467349 DOI: 10.1021/acs.nanolett.2c00596] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Amyloid peptide (AP) self-assembly is a hierarchical process. However, the mechanistic rule of guiding peptides to organize well-ordered nanostructure in a clear and precise manner remains poorly understood. Herein we explored the molecular insight of AP motif aggregates underlying hierarchical process with helical fibrillar structure by atomic force microscope, cryo-electron microscopy (cryo-EM), and molecular dynamics simulation. AP assembly encompasses well-ordered twisted fibrils with uniform morphology, size, and periodicity. More importantly, a heterozipper β-sheet was identified in a protofilament of AP assembly determined by cryo-EM with a high resolution of 3.5 Å. Each peptide heterozipper was further composed of two antiparallel β strands and arranged by an alternative manner in a protofilament. The hydrophobic core and hydrophilic area in each zipper played the significant role for peptide assembling. This work proposed and verified the rule facilitating the basic building unit to form twisted fibrils and gave the explanation of peptide hierarchical assembling.
Collapse
Affiliation(s)
- Yongxiu Song
- Institute for Advanced Materials, Jiangsu University, Zhenjiang, 212013, China
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, 8000, Denmark
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Bin Dai
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Wang
- The Provincial International Science and Technology Cooperation Base on Engineering Biology, International Campus of Zhejiang University, College of Life Sciences, Shanghai Institute for Advanced Study, Institute of Quantitative Biology, Zhejiang University, Haining, 314400, China
| | - Yin Wang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, 8000, Denmark
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 200032, Shanghai, China
| | - Pontus Gourdon
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Department of Experimental Medical Science, Lund University, Lund, 22363, Sweden
| | - Lei Liu
- Institute for Advanced Materials, Jiangsu University, Zhenjiang, 212013, China
| | - Kaituo Wang
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, 8000, Denmark
| |
Collapse
|
46
|
Mukherjee P, Patino CA, Pathak N, Lemaitre V, Espinosa HD. Deep Learning-Assisted Automated Single Cell Electroporation Platform for Effective Genetic Manipulation of Hard-to-Transfect Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107795. [PMID: 35315229 PMCID: PMC9119920 DOI: 10.1002/smll.202107795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/03/2022] [Indexed: 05/03/2023]
Abstract
Genome engineering of cells using CRISPR/Cas systems has opened new avenues for pharmacological screening and investigating the molecular mechanisms of disease. A critical step in many such studies is the intracellular delivery of the gene editing machinery and the subsequent manipulation of cells. However, these workflows often involve processes such as bulk electroporation for intracellular delivery and fluorescence activated cell sorting for cell isolation that can be harsh to sensitive cell types such as human-induced pluripotent stem cells (hiPSCs). This often leads to poor viability and low overall efficacy, requiring the use of large starting samples. In this work, a fully automated version of the nanofountain probe electroporation (NFP-E) system, a nanopipette-based single-cell electroporation method is presented that provides superior cell viability and efficiency compared to traditional methods. The automated system utilizes a deep convolutional network to identify cell locations and a cell-nanopipette contact algorithm to position the nanopipette over each cell for the application of electroporation pulses. The automated NFP-E is combined with microconfinement arrays for cell isolation to demonstrate a workflow that can be used for CRISPR/Cas9 gene editing and cell tracking with potential applications in screening studies and isogenic cell line generation.
Collapse
Affiliation(s)
- Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
- iNfinitesimal LLC, Skokie, IL, 60077, USA
| | - Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- iNfinitesimal LLC, Skokie, IL, 60077, USA
| | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | | | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
- iNfinitesimal LLC, Skokie, IL, 60077, USA
| |
Collapse
|
47
|
Gao L, Xie Z, Zheng M. A general carbon dot-based platform for intracellular delivery of proteins. SOFT MATTER 2022; 18:2776-2781. [PMID: 35315855 DOI: 10.1039/d2sm00204c] [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
The shortcomings of proteins, such as poor stability in biological environments, the impermeability of the membrane and the susceptibility to enzymolysis, restrict their potential applications. Therefore, constructing universal nanocarriers for intracellular delivery of a variety of proteins remains a great challenge. In this work, gallic acid (GA) and L-lysine were used as starting materials to synthesize carbon dots (CDs). The CDs were used as carriers to interact with bovine serum albumin (BSA), enhanced green fluorescent protein (EGFP) and glucose oxidase (GOx) via supramolecular interaction to construct CDs-protein nanocomposites CDs-BSA, CDs-EGFP and CDs-GOx. Furthermore, CDs-EGFP and CDs-GOx can achieve intracellular protein delivery and maintain 89% of the biological activity of GOx. In this work, the latency of CDs is projected as a universal platform for effective intracellular delivery of proteins.
Collapse
Affiliation(s)
- Libo Gao
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun, Jilin 130012, P. R. China.
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Min Zheng
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun, Jilin 130012, P. R. China.
| |
Collapse
|
48
|
Ren Q, Chen Q, Ren L, Cao C, Liu R, Cheng Y. Amphipathic poly-β-peptides for intracellular protein delivery. Chem Commun (Camb) 2022; 58:4320-4323. [PMID: 35293911 DOI: 10.1039/d2cc00453d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of amphipathic poly-β-peptides are designed for intracellular protein delivery. The poly-β-peptides with higher molecular weight and hydrophobic contents exhibit higher protein loading and superior delivery efficiency. The lead material efficiently delivers proteins into cells with reserved bioactivity.
Collapse
Affiliation(s)
- Qianyi Ren
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China.
| | - Qi Chen
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Lanfang Ren
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China.
| | - Chuntao Cao
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China.
| |
Collapse
|
49
|
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
|
50
|
Sun Y, Lau SY, Lim ZW, Chang SC, Ghadessy F, Partridge A, Miserez A. Phase-separating peptides for direct cytosolic delivery and redox-activated release of macromolecular therapeutics. Nat Chem 2022; 14:274-283. [PMID: 35115657 DOI: 10.1038/s41557-021-00854-4] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022]
Abstract
Biomacromolecules are highly promising therapeutic modalities to treat various diseases. However, they suffer from poor cellular membrane permeability, limiting their access to intracellular targets. Strategies to overcome this challenge often employ nanoscale carriers that can get trapped in endosomal compartments. Here we report conjugated peptides that form pH- and redox-responsive coacervate microdroplets by liquid-liquid phase separation that readily cross the cell membrane. A wide range of macromolecules can be quickly recruited within the microdroplets, including small peptides, enzymes as large as 430 kDa and messenger RNAs (mRNAs). The therapeutic-loaded coacervates bypass classical endocytic pathways to enter the cytosol, where they undergo glutathione-mediated release of payload, the bioactivity of which is retained in the cell, while mRNAs exhibit a high transfection efficiency. These peptide coacervates represent a promising platform for the intracellular delivery of a large palette of macromolecular therapeutics that have potential for treating various pathologies (for example, cancers and metabolic diseases) or as carriers for mRNA-based vaccines.
Collapse
Affiliation(s)
- Yue Sun
- Biological and Biomimetic Material Laboratory (BBML), Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore, Singapore
| | - Sze Yi Lau
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), Neuros/Immunos, Singapore, Singapore
| | - Zhi Wei Lim
- Biological and Biomimetic Material Laboratory (BBML), Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore, Singapore
| | - Shi Chieh Chang
- Translation Medicine Research Centre, MSD International, Singapore, Singapore
| | - Farid Ghadessy
- p53 Laboratory, Agency for Science, Technology and Research (A*STAR), Neuros/Immunos, Singapore, Singapore
| | - Anthony Partridge
- Translation Medicine Research Centre, MSD International, Singapore, Singapore
| | - Ali Miserez
- Biological and Biomimetic Material Laboratory (BBML), Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore, Singapore. .,School of Biological Sciences, NTU, Singapore, Singapore.
| |
Collapse
|