1
|
Lee YJ, Hong J, Seo BY, Lee BH, Sarangthem V, Park RW. Strategic Optimization of Nanoparticle Characteristics to Enhance Tumor Targeting and Doxorubicin Delivery. Int J Nanomedicine 2025; 20:6357-6378. [PMID: 40416731 PMCID: PMC12103874 DOI: 10.2147/ijn.s513336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 05/12/2025] [Indexed: 05/27/2025] Open
Abstract
Background Doxorubicin (Dox) is a potent anticancer agent; however, its therapeutic efficacy is constrained by a narrow therapeutic index, resulting in nonselective cardiotoxicity and nephrotoxicity. To improve its specificity and therapeutic efficacy, multivalent targeting strategies are being developed. Methods A chimeric polypeptide consisting of an elastin-like polypeptides (ELP) copolymer with a repeating IL-4 receptor-specific targeting peptide, AP-1, and a (GGCGSCGSC)2 sequence encoding 6 cysteine residues (C6) at the carboxyl-terminus for Dox conjugation was designed. Several AP1-ELPs of varying molecular sizes and structures, ranging from unimers to micelle-forming polymers, were characterized to evaluate their influence on Dox delivery and tumor inhibition. Results Conjugating Dox to the C6 via an acid-labile linker induced self-assembly into micelle-like structures at body temperature. The size of these multivalent constructs significantly influenced their tumor penetration and overall therapeutic outcomes. High molecular weight, micelle-forming AP1-ELP constructs demonstrated faster tumor entry and enhanced inhibition compared to lower molecular weight linear AP1-ELPs. Tumor uptake of Dox was five times greater than that of free drug and twice that of low molecular weight, linear AP1-ELPs. Furthermore, systemic administration of these high molecular weight constructs effectively inhibited tumor growth in breast carcinoma xenograft models without inducing specific organ toxicity. Conclusion Outperforming free Dox, high molecular weight micelle-forming AP1-ELP constructs achieve superior tumor targeting and efficacy with minimal toxicity, highlighting their potential as safer and more promising carriers for targeted drug delivery.
Collapse
Affiliation(s)
- Young-Jin Lee
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Jisan Hong
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Bo-Yeon Seo
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Byung-Heon Lee
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Vijaya Sarangthem
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, Cell & Matrix Research Institute, Kyungpook National University, School of Medicine, Daegu, 41944, Republic of Korea
| |
Collapse
|
2
|
Gong L, Qi Y, Zhang F, Sun Y, Wang X, Gao W. Thermo-pH-Sensitive Polypeptides Boost Protein Drug's Tumor Permeation and Pharmacology. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2501787. [PMID: 40143732 DOI: 10.1002/smll.202501787] [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] [Received: 02/16/2025] [Revised: 03/12/2025] [Indexed: 03/28/2025]
Abstract
Proteins are typically subject to poor stability, short half-life, and poor cell and tissue permeability, which restrict their wide applications as drugs for disease treatment. Current protein modification techniques mostly focus on improving the stability and half-life of proteins, but hardly solve their poor cell and tissue permeability. To address this issue, the study innovatively designs thermo-pH-sensitive elastin-like polypeptides to modify proteins, named ELP(HX)n in which histidine (H) and any amino acid except proline (X) are guest amino acids in the polypeptides and n is the total number of the guest amino acids. H in ELP(HX)n can be protonated under acidic conditions. To prove the concept, an important protein drug of L-asparaginase (ASP) is genetically fused to ELP(HV)60 to generate ASP-ELP(HV)60. Compared with ASP and PEGylated ASP, ASP-ELP(HV)60 exhibits not only elevated stability and extended half-life but also enhanced tumor cell and tissue penetration, resulting in improved antitumor efficacy. These findings demonstrate that ELP(HX)n fusion is a novel and general protein modification method to overcome the intrinsic limitations of proteins as therapeutics, rendering it feasible to design intelligent protein therapeutics, especially for efficient tumor therapy.
Collapse
Affiliation(s)
- Like Gong
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Yanshuang Qi
- Biomedical Engineering, Department of Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Fan Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
| | - Yuanzi Sun
- Biomedical Engineering, Department of Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Xuan Wang
- Biomedical Engineering, Department of Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Weiping Gao
- Institute of Medical Technology, Biomedical Engineering Department of Institute of Advanced Clinical Medicine, Peking University-Yunnan Baiyao International Medical Research Center, Frontiers Science Center for Cancer Integrative Omics of Peking University International Cancer Institute, State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Department of Biomedical Engineering in College of Future Technology, Peking University, Beijing, 100191, China
| |
Collapse
|
3
|
Chowdhury S, Beeton K, Wallace Z, Moore M, Bidwell GL, Janorkar AV. Extending the Three-Dimensional Culture of Adipocytes Through Surface Coatings. Bioengineering (Basel) 2025; 12:266. [PMID: 40150730 PMCID: PMC11939550 DOI: 10.3390/bioengineering12030266] [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: 01/27/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
To mimic the important features of progressing adiposity, in vitro adipose cell culture models must allow gradual intracellular fat accumulation in the three-dimensional (3D) arrangement of adipose-derived stem cells (ASCs) over a long-term culture period. Previously, elastin-like polypeptide (ELP) and polyethyleneimine (PEI) have been used to culture human adipose-derived stem cells (hASCs) as 3D spheroids and to differentiate them to adipocytes over a relatively long culture period of up to 5 weeks. In this study, to further enhance the spheroid adhesion properties, ELP was fused with Arginine-Glycine-Aspartic Acid (RGD) residues, known for their role as cell-attachment sites. This study aimed to assess whether the addition of RGD to the C-or N-terminus of ELP would impact the spheroid-forming ability of ELP-PEI coatings. ELP-RGD conjugates were produced using genetically modified Escherichia coli to express ELP-(RGD)3 and (RGD)3-ELP, followed by chemical conjugation with PEI. SDS gel electrophoresis, FTIR spectroscopy, and turbidimetry analyses revealed that ELP was conjugated with RGD without much alteration in the molecular weight, functional groups present, and transition temperature of ELP. The addition of RGD to ELP also did not affect the chemical conjugation capacity of ELP to PEI. We observed that the ELP-PEI coating formed slightly larger spheroids (61.8 ± 3.2 µm) compared to the ELP-(RGD)3-PEI and (RGD)3-ELP-PEI coatings (56.6 ± 3.0 and 53.4 ± 2.4 µm, respectively). Despite the size difference, ELP-(RGD)3-PEI coatings exhibited superior spheroid retention during media changes, with minimal spheroid loss. DNA assay results confirmed a significant decrease in the DNA concentration (p < 0.05) after the 20 media changes for spheroids cultured on the ELP-PEI coating, indicating spheroid loss. However, there was no significant difference in DNA concentration before and after 20 media changes for spheroids cultured on the ELP-(RGD)3-PEI and (RGD)3-ELP-PEI coatings (p > 0.05). These findings suggest that RGD incorporation does not hinder the initial spheroid formation ability of the ELP-PEI coating and enhances spheroid retention under dynamic culture conditions.
Collapse
Affiliation(s)
- Sheetal Chowdhury
- Department of Biomedical Materials Science, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Komal Beeton
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Zacchaeus Wallace
- Department of Biomedical Materials Science, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Maggie Moore
- Department of Biomedical Materials Science, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Amol V. Janorkar
- Department of Biomedical Materials Science, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| |
Collapse
|
4
|
Varanko AK, Deshpande S, Li X, Chilkoti A. Binding Strength, Not Valency, Dictates Accumulation and Penetration of Affinity Targeted Macromolecules into Solid Tumors. Biomacromolecules 2025; 26:503-513. [PMID: 39729341 DOI: 10.1021/acs.biomac.4c01303] [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: 12/28/2024]
Abstract
The efficacy of tumor-targeted therapeutics, engineered to engage specific cellular receptors to promote accumulation and penetration, is strongly influenced by the carrier's affinity for its target and the valency of binding molecules incorporated into the carrier. Previous research has primarily focused on improving targeting by augmenting the number of binding proteins on the carrier, inadvertently raising avidity without isolating the individual effects of binding strength and valency. Herein, we precisely evaluate the impact of multivalency on tumor targeting with a recombinant approach to independently control valency, avidity, and size. Our findings reveal that constructs with equivalent binding strength exhibit comparable receptor engagement and tumor extravasation, regardless of valency. Moreover, excessive avidity adversely affected tumor accumulation and penetration, with the highest-avidity construct showing diminished exposure. These results indicate that overall binding strength, not valency, is the primary determinant of tumor targeting, providing valuable insights for designing effective macromolecular drug carriers.
Collapse
Affiliation(s)
- Anastasia K Varanko
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Sonal Deshpande
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Xinghai Li
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
5
|
Mondal M, Jankoski PE, Lee LD, Dinakarapandian DM, Chiu TY, Swetman WS, Wu H, Paravastu AK, Clemons TD, Rangachari V. Reversible Disulfide Bond Cross-Links as Tunable Levers of Phase Separation in Designer Biomolecular Condensates. J Am Chem Soc 2024; 146:25299-25311. [PMID: 39196681 PMCID: PMC11403603 DOI: 10.1021/jacs.4c09557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Biomolecular condensates (BCs) are membraneless hubs enriched with proteins and nucleic acids that have emerged as important players in many cellular functions. Uncovering the sequence determinants of proteins for phase separation is essential in understanding the biophysical and biochemical properties of BCs. Despite significant discoveries in the past decade, the role of cysteine residues in BC formation and dissolution has remained unknown. Here, to uncover the involvement of disulfide cross-links and their redox sensitivity in BCs, we designed a "stickers and spacers" model of phase-separating peptides interspersed with cysteines. Through biophysical investigations, we learned that cysteines promote liquid-liquid phase separation in oxidizing conditions and perpetuate liquid condensates through disulfide cross-links, which can be reversibly tuned with redox chemistry. By varying the composition of cysteines, subtle but distinct changes in the viscoelastic behavior of the condensates were observed. Empirically, we conclude that cysteines function neither as stickers nor spacers but as covalent nodes to lower the effective concentrations for sticker interactions and inhibit system-spanning percolation networks. Together, we unmask the possible role of cysteines in the formation of biomolecular condensates and their potential use as tunable covalent cross-linkers in developing redox-sensitive viscoelastic materials.
Collapse
Affiliation(s)
- Malay Mondal
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Penelope E Jankoski
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Landon D Lee
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Daniel M Dinakarapandian
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0002, United States
| | - Tzu-Ying Chiu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0002, United States
| | - Windfield S Swetman
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Hongwei Wu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0002, United States
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0002, United States
| | - Tristan D Clemons
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| | - Vijayaraghavan Rangachari
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406, United States
| |
Collapse
|
6
|
Ali A, Phan A, Vaikari V, Park M, Pospiech M, Chu R, Meng Y, MacKay JA, Alachkar H. FLT3/CD99 Bispecific Antibody-Based Nanoparticles for Acute Myeloid Leukemia. CANCER RESEARCH COMMUNICATIONS 2024; 4:1946-1962. [PMID: 39007347 PMCID: PMC11305399 DOI: 10.1158/2767-9764.crc-24-0096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/22/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Cluster of differentiation 99 (CD99) is a receptor that is significantly upregulated in acute myeloid leukemia (AML). FMS-like tyrosine kinase 3 internal tandem duplication mutation in AML (FLT3-ITD AML) exhibits even higher levels of CD99 expression. Our group previously employed a novel peptide platform technology called elastin-like polypeptides and fused it with single-chain antibodies capable of binding to FLT3 (FLT3-A192) or CD99 (CD99-A192). Targeting either FLT3 or CD99 using FLT3-A192 or CD99-A192 led to AML cell death and reduced leukemia burden in AML mouse models. Here, we report on the development of a novel Co-Assembled construct that is capable of binding to both CD99 and FLT3 and the antileukemia activity of the bispecific construct in FLT3-ITD AML preclinical models. This dual-targeting Co-Assembled formulation exhibits cytotoxic effects on AML cells (AML cell lines and primary blasts) and reduced leukemia burden and prolonged survival in FLT3-ITD AML mouse models. Altogether, this study demonstrates the potential of an innovative therapeutic strategy that targets both FLT3 and CD99 in FLT3-ITD AML. SIGNIFICANCE This study investigates a dual-targeting strategy in acute myeloid leukemia (AML), focusing on FLT3 and CD99. The approach demonstrates enhanced therapeutic potential, presenting a novel option for AML treatment.
Collapse
Affiliation(s)
- Atham Ali
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Vijaya Vaikari
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Mincheol Park
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Mateusz Pospiech
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Ryan Chu
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Yiting Meng
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, University of Southern California, Los Angeles, California.
- Department of Ophthalmology, USC Roski Eye Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California.
- Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California.
| | - Houda Alachkar
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.
| |
Collapse
|
7
|
Mondal M, Jankoski PE, Lee LD, Dinakarapandian DM, Chiu TY, Swetman WS, Wu H, Paravastu AK, Clemons TD, Rangachari V. Reversible disulfide bond crosslinks as tunable levers of phase separation in designer biomolecular condensates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603402. [PMID: 39071339 PMCID: PMC11275914 DOI: 10.1101/2024.07.13.603402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Biomolecular condensates (BCs) are membraneless hubs enriched in proteins and nucleic acids that have become important players in many cellular functions. Uncovering the sequence determinants of proteins for phase separation is important in understanding the biophysical and biochemical properties of BCs. Despite significant discoveries in the last decade, the role of cysteine residues in BC formation and dissolution has remained unknown. Here, to determine the involvement of disulfide crosslinks and their redox sensitivity in BCs, we designed a 'stickers and spacers' model of phase-separating peptides interspersed with cysteines. Through biophysical investigations, we learned that cysteines promote liquid-liquid phase separation in oxidizing conditions and perpetuate liquid condensates through disulfide crosslinks, which can be reversibly tuned with redox chemistry. By varying the composition of cysteines, subtle but distinct changes in the viscoelastic behavior of the condensates were observed. Empirically, we conclude that cysteines are neither stickers nor spacers but function as covalent nodes to lower the effective concentrations for sticker interactions and inhibit system-spanning percolation networks. Together, we unmask the role of cysteines in protein phase behavior and the potential to develop tunable, redox-sensitive viscoelastic materials.
Collapse
|
8
|
Cecuda-Adamczewska V, Romanik-Chruścielewska A, Kosowska K, Sokołowska I, Łukasiewicz N, Korycka P, Florys-Jankowska K, Zakrzewska A, Wszoła M, Klak M. Elasticity Modification of Biomaterials Used in 3D Printing with an Elastin-Silk-like Recombinant Protein. J Funct Biomater 2024; 15:141. [PMID: 38921515 PMCID: PMC11204424 DOI: 10.3390/jfb15060141] [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: 03/28/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
The recombinant structural protein described in this study was designed based on sequences derived from elastin and silk. Silk-elastin hybrid copolymers are characterized by high solubility while maintaining high product flexibility. The phase transition temperature from aqueous solution to hydrogel, as well as other physicochemical and mechanical properties of such particles, can differ significantly depending on the number of sequence repeats. We present a preliminary characterization of the EJ17zipR protein obtained in high yield in a prokaryotic expression system and efficiently purified via a multistep process. Its addition significantly improves biomaterial's rheological and mechanical properties, especially elasticity. As a result, EJ17zipR appears to be a promising component for bioinks designed to print spatially complex structures that positively influence both shape retention and the internal transport of body fluids. The results of biological studies indicate that the addition of the studied protein creates a favorable microenvironment for cell adhesion, growth, and migration.
Collapse
Affiliation(s)
- Violetta Cecuda-Adamczewska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | | | - Katarzyna Kosowska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Iwona Sokołowska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Natalia Łukasiewicz
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Paulina Korycka
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Katarzyna Florys-Jankowska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | | | - Michał Wszoła
- Polbionica Ltd., 01-424 Warsaw, Poland; (A.Z.); (M.W.)
| | - Marta Klak
- Polbionica Ltd., 01-424 Warsaw, Poland; (A.Z.); (M.W.)
| |
Collapse
|
9
|
Levêque M, Lecommandoux S, Garanger E. Thermoresponsive Core-cross-linked Nanoparticles from HA- b-ELP Diblock Copolymers. Biomacromolecules 2024; 25:3011-3017. [PMID: 38689515 DOI: 10.1021/acs.biomac.4c00137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Stabilization against the dilution-dependent disassembly of self-assembled nanoparticles is a requirement for in vivo application. Herein, we propose a simple and biocompatible cross-linking reaction for the stabilization of a series of nanoparticles formed by the self-assembly of amphiphilic HA-b-ELP block copolymers, through the alkylation of methionine residues from the ELP block with diglycidyl ether compounds. The core-cross-linked nanoparticles retain their colloidal properties, with a spherical core-shell morphology, while maintaining thermoresponsive behavior. As such, instead of a reversible disassembly when non-cross-linked, a reversible swelling of nanoparticles' core and increase of hydrodynamic diameter are observed with lowering of the temperature.
Collapse
Affiliation(s)
- Manon Levêque
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France
| | | | - Elisabeth Garanger
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France
| |
Collapse
|
10
|
Walter V, Schmatko T, Muller P, Schroder AP, MacEwan SR, Chilkoti A, Marques CM. Negative lipid membranes enhance the adsorption of TAT-decorated elastin-like polypeptide micelles. Biophys J 2024; 123:901-908. [PMID: 38449310 PMCID: PMC10995422 DOI: 10.1016/j.bpj.2024.03.001] [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: 11/20/2023] [Revised: 02/07/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
A cell-penetrating peptide (CPP) is a short amino-acid sequence capable of efficiently translocating across the cellular membrane of mammalian cells. However, the potential of CPPs as a delivery vector is hampered by the strong reduction of its translocation efficiency when it bears an attached molecular cargo. To overcome this problem, we used previously developed diblock copolymers of elastin-like polypeptides (ELPBCs), which we end functionalized with TAT (transactivator of transcription), an archetypal CPP built from a positively charged amino acid sequence of the HIV-1 virus. These ELPBCs self-assemble into micelles at a specific temperature and present the TAT peptide on their corona. These micelles can recover the lost membrane affinity of TAT and can trigger interactions with the membrane despite the presence of a molecular cargo. Herein, we study the influence of membrane surface charge on the adsorption of TAT-functionalized ELP micelles onto giant unilamellar vesicles (GUVs). We show that the TAT-ELPBC micelles show an increased binding constant toward negatively charged membranes compared to neutral membranes, but no translocation is observed. The affinity of the TAT-ELPBC micelles for the GUVs displays a stepwise dependence on the lipid charge of the GUV, which, to our knowledge, has not been reported previously for interactions between peptides and lipid membranes. By unveiling the key steps controlling the interaction of an archetypal CPP with lipid membranes, through regulation of the charge of the lipid bilayer, our results pave the way for a better design of delivery vectors based on CPPs.
Collapse
Affiliation(s)
- Vivien Walter
- Institut Charles Sadron, CNRS UPR22 & Université de Strasbourg, Strasbourg, France
| | - Tatiana Schmatko
- Institut Charles Sadron, CNRS UPR22 & Université de Strasbourg, Strasbourg, France.
| | - Pierre Muller
- Institut Charles Sadron, CNRS UPR22 & Université de Strasbourg, Strasbourg, France
| | | | - Sarah R MacEwan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Carlos M Marques
- University of Lyon, ENS-Lyon, CNRS UMR 5182, Chem. Lab, Lyon, France.
| |
Collapse
|
11
|
Yu S, Chen W, Liu G, Flores B, DeWolf EL, Fan B, Xiang Y, Webber MJ. Glucose-Driven Droplet Formation in Complexes of a Supramolecular Peptide and Therapeutic Protein. J Am Chem Soc 2024; 146:7498-7505. [PMID: 38465595 DOI: 10.1021/jacs.3c13139] [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: 03/12/2024]
Abstract
Biology achieves remarkable function through processes arising from spontaneous or transient liquid-liquid phase separation (LLPS) of proteins and other biomolecules. While polymeric systems can achieve similar phenomena through simple or complex coacervation, LLPS with supramolecular materials has been less commonly shown. Functional applications for synthetic LLPS systems are an expanding area of emphasis, with particular focus on capturing the transient and dynamic state of these structures for use in biomedicine. Here, a net-cationic supramolecular peptide amphiphile building block with a glucose-binding motif is shown that forms LLPS structures when combined with a net-negatively charged therapeutic protein, dasiglucagon, in the presence of glucose. The droplets that arise are dynamic and coalesce quickly. However, the interface can be stabilized by addition of a 4-arm star PEG. When the stabilized droplets formed in glucose are transferred to a bulk phase containing different glucose concentrations, their stability and lifetime decrease according to bulk glucose concentration. This glucose-dependent formation translates into an accelerated release of dasiglucagon in the absence of glucose; this hormone analogue itself functions therapeutically to correct low blood glucose (hypoglycemia). These droplets also offer function in mitigating the most severe effects of hypoglycemia arising from an insulin overdose through delivery of dasiglucagon in a mouse model of hypoglycemic rescue. Accordingly, this approach to use complexation between a supramolecular peptide amphiphile and a therapeutic protein in the presence of glucose leads to droplets with functional potential to dissipate for the release of the therapeutic material in low blood glucose environments.
Collapse
Affiliation(s)
- Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Weike Chen
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Guoqiang Liu
- Integrated Biomedical Sciences Program, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Belen Flores
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Emily L DeWolf
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Bowen Fan
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Yuanhui Xiang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
12
|
Phan A, MacKay JA. Steric stabilization of bioactive nanoparticles using elastin-like polypeptides. Adv Drug Deliv Rev 2024; 206:115189. [PMID: 38281625 PMCID: PMC11580827 DOI: 10.1016/j.addr.2024.115189] [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: 11/28/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Elastin-like polypeptides (ELP) are versatile, thermo-responsive polymers that can be conjugated to virtually any therapeutic cargo. Derived from short amino-acid sequences and abundant in humans, certain ELPs display low immunogenicity. Substrates for endogenous proteases, ELPs are biodegradable and thus, are candidate biomaterials. Peptides and proteins can be directly coupled with ELPs through genetic engineering, while other polymers and small molecules can be appended through covalent bioconjugation or non-covalent complexation. ELPs that phase separate at physiological temperatures can form the core of nano assemblies; however, ELPs that remain soluble can sterically stabilize the corona of a variety of nanoparticles. Nanoparticles with ELPs at their corona promote colloids with favorable pharmacokinetic (PK) properties that enables therapeutic efficacy with intermittent administration. This review highlights a comprehensive spectrum of ELP fusions shown to stabilize the solubility, and sometimes bioactivity, of their cargo - with a focus on biophysical properties that underlie their therapeutic effects.
Collapse
Affiliation(s)
- Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
13
|
Yang N, Sun M, Wang H, Hu D, Zhang A, Khan S, Chen Z, Chen D, Xie S. Progress of stimulus responsive nanosystems for targeting treatment of bacterial infectious diseases. Adv Colloid Interface Sci 2024; 324:103078. [PMID: 38215562 DOI: 10.1016/j.cis.2024.103078] [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: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
In recent decades, due to insufficient concentration at the lesion site, low bioavailability and increasingly serious resistance, antibiotics have become less and less dominant in the treatment of bacterial infectious diseases. It promotes the development of efficient drug delivery systems, and is expected to achieve high absorption, targeted drug release and satisfactory therapy effects. A variety of endogenous stimulation-responsive nanosystems have been constructed by using special infection microenvironments (pH, enzymes, temperature, etc.). In this review, we firstly provide an extensive review of the current research progress in antibiotic treatment dilemmas and drug delivery systems. Then, the mechanism of microenvironment characteristics of bacterial infected lesions was elucidated to provide a strong theoretical basis for bacteria-targeting nanosystems design. In particular, the discussion focuses on the design principles of single-stimulus and dual-stimulus responsive nanosystems, as well as the use of endogenous stimulus-responsive nanosystems to deliver antimicrobial agents to target locations for combating bacterial infectious diseases. Finally, the challenges and prospects of endogenous stimulus-responsive nanosystems were summarized.
Collapse
Affiliation(s)
- Niuniu Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Mengyuan Sun
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Huixin Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Danlei Hu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Aoxue Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Suliman Khan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Zhen Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
14
|
Azulay R, Strugach DS, Amiram M. Self-assembly of temperature-responsive di-block polypeptides functionalized with unnatural amino acids. Protein Sci 2024; 33:e4878. [PMID: 38147468 PMCID: PMC10804675 DOI: 10.1002/pro.4878] [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: 07/18/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
The incorporation of unnatural amino acids (uAAs) into protein-based polymers has emerged as a powerful methodology to expand their chemical repertoire. Recently, we demonstrated that incorporating uAAs into two temperature-responsive protein-based polymers-namely resilin- and elastin-like polypeptides (RLPs and ELPs, respectively)-can alter their properties. In this study, we incorporated aromatic uAAs into the protein sequence of RLP-ELP diblocks to yield new and diverse assemblies from a single DNA template. Specifically, we show that incorporating aromatic uAAs can modulate the phase-transition behaviors and self-assembly of the diblocks into various morphologies, including spherical and cylindrical micelles and single- and double-layered vesicles, with some constructs also demonstrating a temperature-responsive shape-shifting behavior. Next, we evaluated the ability of the RLP-ELP assemblies to encapsulate a chemotherapeutic drug, doxorubicin, and show how the identity of the incorporated uAAs and the morphology of the nanostructure affect the encapsulation efficiency. Taken together, our findings demonstrate that the multi-site incorporation of uAAs into temperature-responsive, amphiphilic protein-based diblock copolymers is a promising approach for the functionalization and tuning of self-assembled nanostructures.
Collapse
Affiliation(s)
- Rotem Azulay
- Avram and Stella Goldstein‐Goren Department of Biotechnology EngineeringBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Daniela S. Strugach
- Avram and Stella Goldstein‐Goren Department of Biotechnology EngineeringBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Miriam Amiram
- Avram and Stella Goldstein‐Goren Department of Biotechnology EngineeringBen‐Gurion University of the NegevBeer‐ShevaIsrael
| |
Collapse
|
15
|
Kuperkar K, Atanase LI, Bahadur A, Crivei IC, Bahadur P. Degradable Polymeric Bio(nano)materials and Their Biomedical Applications: A Comprehensive Overview and Recent Updates. Polymers (Basel) 2024; 16:206. [PMID: 38257005 PMCID: PMC10818796 DOI: 10.3390/polym16020206] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Degradable polymers (both biomacromolecules and several synthetic polymers) for biomedical applications have been promising very much in the recent past due to their low cost, biocompatibility, flexibility, and minimal side effects. Here, we present an overview with updated information on natural and synthetic degradable polymers where a brief account on different polysaccharides, proteins, and synthetic polymers viz. polyesters/polyamino acids/polyanhydrides/polyphosphazenes/polyurethanes relevant to biomedical applications has been provided. The various approaches for the transformation of these polymers by physical/chemical means viz. cross-linking, as polyblends, nanocomposites/hybrid composites, interpenetrating complexes, interpolymer/polyion complexes, functionalization, polymer conjugates, and block and graft copolymers, are described. The degradation mechanism, drug loading profiles, and toxicological aspects of polymeric nanoparticles formed are also defined. Biomedical applications of these degradable polymer-based biomaterials in and as wound dressing/healing, biosensors, drug delivery systems, tissue engineering, and regenerative medicine, etc., are highlighted. In addition, the use of such nano systems to solve current drug delivery problems is briefly reviewed.
Collapse
Affiliation(s)
- Ketan Kuperkar
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology (SVNIT), Ichchhanath, Piplod, Surat 395007, Gujarat, India;
| | - Leonard Ionut Atanase
- Faculty of Medical Dentistry, “Apollonia” University of Iasi, 700511 Iasi, Romania
- Academy of Romanian Scientists, 050045 Bucharest, Romania
| | - Anita Bahadur
- Department of Zoology, Sir PT Sarvajanik College of Science, Surat 395001, Gujarat, India;
| | - Ioana Cristina Crivei
- Department of Public Health, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” University of Life Sciences, 700449 Iasi, Romania;
| | - Pratap Bahadur
- Department of Chemistry, Veer Narmad South Gujarat University (VNSGU), Udhana-Magdalla Road, Surat 395007, Gujarat, India;
| |
Collapse
|
16
|
Bhardwaj R, Mishra P. Multiresponsive Nanoscale Self-Assembly of Azurin-Elastin-like Polypeptide Fusion Protein for Enhanced Prostate Cancer Therapy. Biomacromolecules 2024; 25:508-521. [PMID: 38047916 DOI: 10.1021/acs.biomac.3c01125] [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: 12/05/2023]
Abstract
A fusion protein composed of a bacterial protein, azurin, having antineoplastic properties and a thermally responsive structural cationic elastin-like protein (ELP), is designed, cloned, expressed, and purified. A simple method of inverse transition cycle (ITC) is employed to purify the fusion protein azurin-ELP diblock copolymer (d-bc). The molecular weight of the azurin-ELP fusion protein is ∼32 kDa. Further, its self-assembly properties are investigated. Interestingly, the engineered azurin-ELP d-bc in response to increasing temperature shows a dual-step phase separation into biofunctional nanostructures. Around the physiological temperature, azurin-ELP d-bc forms stable coacervates, which is dependent on the concentration and time of incubation. These coacervates are formed below the lower critical solubility temperature (LCST) of the ELP block at physiological temperature. Above LCST, i.e., 50-55°C, micelles of size ranging from 25 to 30 nm are formed. The cytotoxicity of azurin-ELP d-bc depends on the size of the coacervates formed and their cellular uptake at physiological temperature. Further, MTT assay of azurin-ELP d-bc in the cross-linked micelles prepared ex situ shows > six times higher killing of LNCaP cells than the unimeric form of azurin-ELP at 5 μM concentration. The flow cytometric results of these micelles at 20 μM concentration show ∼97% LNCaP cells in the apoptotic phase. Thus, azurin-ELP cross-linked micelles have enhanced potential for anticancer therapy due to their higher avidity.
Collapse
Affiliation(s)
- Ritu Bhardwaj
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Prashant Mishra
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
17
|
Chen H, Zhang Q. Polypeptides as alternatives to PEGylation of therapeutic agents. Expert Opin Drug Deliv 2024; 21:1-12. [PMID: 38116624 DOI: 10.1080/17425247.2023.2297937] [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: 10/05/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Due to the concerns raised by the extensive application of PEGylation, polypeptides have stood out as excellent candidates with adequate biocompatibility and biodegradability with tunable hydrophilicity. AREAS COVERED In this review, polypeptides with the potential to replace PEGylation have been summarized and their application has been reviewed, including XTEN, PASylation, polysarcosine, zwitterion polypeptides, ELPylation, etc. Besides their strengths, the remaining challenges have also been discussed and the future perspectives have been provided. EXPERT OPINION Polypeptides have been applied in the designing of peptide/protein drugs as well as nanomedicines, and some of the pharmaceutics have made it into the clinical trials and got approved. These polypeptides showed similar hydrophilic properties to PEGylation, which increased the hydrodynamic volumes of protein drugs, reduced kidney elimination, decreased protein-polymer interaction and potentially improved the drug delivery efficiency due to the extended circulation time in the system. Moreover, they demonstrated superior biodegradability and biocompatibility, compensating for the deficiencies for polymers such as PEG.
Collapse
Affiliation(s)
- Huali Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Qianyu Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Luu CH, Nguyen N, Ta HT. Unravelling Surface Modification Strategies for Preventing Medical Device-Induced Thrombosis. Adv Healthc Mater 2024; 13:e2301039. [PMID: 37725037 PMCID: PMC11468451 DOI: 10.1002/adhm.202301039] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/29/2023] [Indexed: 09/21/2023]
Abstract
The use of biomaterials in implanted medical devices remains hampered by platelet adhesion and blood coagulation. Thrombus formation is a prevalent cause of failure of these blood-contacting devices. Although systemic anticoagulant can be used to support materials and devices with poor blood compatibility, its negative effects such as an increased chance of bleeding, make materials with superior hemocompatibility extremely attractive, especially for long-term applications. This review examines blood-surface interactions, the pathogenesis of clotting on blood-contacting medical devices, popular surface modification techniques, mechanisms of action of anticoagulant coatings, and discusses future directions in biomaterial research for preventing thrombosis. In addition, this paper comprehensively reviews several novel methods that either entirely prevent interaction between material surfaces and blood components or regulate the reaction of the coagulation cascade, thrombocytes, and leukocytes.
Collapse
Affiliation(s)
- Cuong Hung Luu
- School of Environment and ScienceGriffith UniversityNathanQueensland4111Australia
- Queensland Micro‐ and Nanotechnology CentreGriffith UniversityNathanQueensland4111Australia
| | - Nam‐Trung Nguyen
- School of Environment and ScienceGriffith UniversityNathanQueensland4111Australia
- Queensland Micro‐ and Nanotechnology CentreGriffith UniversityNathanQueensland4111Australia
| | - Hang Thu Ta
- School of Environment and ScienceGriffith UniversityNathanQueensland4111Australia
- Queensland Micro‐ and Nanotechnology CentreGriffith UniversityNathanQueensland4111Australia
| |
Collapse
|
19
|
Grazon C, Garanger E, Lalanne P, Ibarboure E, Galagan JE, Grinstaff MW, Lecommandoux S. Transcription-Factor-Induced Aggregation of Biomimetic Oligonucleotide- b-Protein Micelles. Biomacromolecules 2023; 24:5027-5034. [PMID: 37877162 DOI: 10.1021/acs.biomac.3c00662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Polymeric micelles and especially those based on natural diblocks are of particular interest due to their advantageous properties in terms of molecular recognition, biocompatibility, and biodegradability. We herein report a facile and straightforward synthesis of thermoresponsive elastin-like polypeptide (ELP) and oligonucleotide (ON) diblock bioconjugates, ON-b-ELP, through copper-catalyzed azide-alkyne cycloaddition. The resulting thermosensitive diblock copolymer self-assembles above its critical micelle temperature (CMT ∼30 °C) to form colloidally stable micelles of ∼50 nm diameter. The ON-b-ELP micelles hybridize with an ON complementary strand and maintain their size and stability. Next, we describe the capacity of these micelles to bind proteins, creating more complex structures using the classic biotin-streptavidin pairing and the specific recognition between a transcription factor protein and the ON strand. In both instances, the micelles are intact, form larger structures, and retain their sensitivity to temperature.
Collapse
Affiliation(s)
- Chloé Grazon
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, Talence F-33400, France
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Elisabeth Garanger
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France
| | - Pierre Lalanne
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France
| | - Emmanuel Ibarboure
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France
| | - James E Galagan
- Department of Microbiology, Boston University, Boston, Massachusetts 02118, United States
| | - Mark W Grinstaff
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | | |
Collapse
|
20
|
Roland TJ, Strauss GL, Bushra N, Muschol M, Koria P. Isoelectric point (pI)-based phase separation (pI-BPS) purification of elastin-like polypeptides (ELPs) containing charged, biologically active fusion proteins (ELP-FPs). Biotechnol Prog 2023; 39:e3381. [PMID: 37531360 DOI: 10.1002/btpr.3381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/07/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Elastin-like polypeptides (ELPs) are peptide-based biomaterials with residue sequence (VPGXG)n where X is any residue except proline. ELPs are a useful modality for delivering biologically active proteins (growth factors, protease inhibitors, anti-inflammatory peptides, etc.) as fusion proteins (ELP-FP). ELP-FPs are particularly cost-effective because they can be rapidly purified using Inverse Temperature Cycling (ITC) via the reversible formation and precipitation of entropically driven aggregates above a transition temperature (Tt ). When ELP fusion proteins (ELP-FPs) contain significant charge density at physiological pH, electrostatic repulsion between them severely inhibits aggregate formation. The literature does not currently describe methods for purifying ELP-FPs containing charged proteins on either side of the ELP sequence as fusion partners without organic solvents. Here, the isoelectric point (pI) of ELP-FPs is discussed as a means of neutralizing surface charges on ELP-FPs and increasing ITC yield to dramatically high levels. We use pI-based phase separation (pI-BPS) to purify ELP-FPs containing cationic and anionic fusion proteins. We report a dramatic increase in protein yield when using pI-BPS for purification of ELP-FPs. Proteins purified by this method also retain the functional activity of the protein present in the ELP-FP. Techniques developed here enable significant diversification of possible fusion proteins delivered by ELPs as ELP-FPs by allowing them to be produced and purified at higher quantities and yields.
Collapse
Affiliation(s)
- Truman J Roland
- Department of Chemical, Biological & Materials Engineering, University of South Florida, Tampa, Florida, USA
| | - Graham L Strauss
- Department of Chemical, Biological & Materials Engineering, University of South Florida, Tampa, Florida, USA
| | - Nabila Bushra
- Department of Physics, University of South Florida, Tampa, Florida, USA
| | - Martin Muschol
- Department of Physics, University of South Florida, Tampa, Florida, USA
| | - Piyush Koria
- Department of Chemical, Biological & Materials Engineering, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
21
|
Kaltbeitzel J, Wich PR. Protein-based Nanoparticles: From Drug Delivery to Imaging, Nanocatalysis and Protein Therapy. Angew Chem Int Ed Engl 2023; 62:e202216097. [PMID: 36917017 DOI: 10.1002/anie.202216097] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/16/2023]
Abstract
Proteins and enzymes are versatile biomaterials for a wide range of medical applications due to their high specificity for receptors and substrates, high degradability, low toxicity, and overall good biocompatibility. Protein nanoparticles are formed by the arrangement of several native or modified proteins into nanometer-sized assemblies. In this review, we will focus on artificial nanoparticle systems, where proteins are the main structural element and not just an encapsulated payload. While under natural conditions, only certain proteins form defined aggregates and nanoparticles, chemical modifications or a change in the physical environment can further extend the pool of available building blocks. This allows the assembly of many globular proteins and even enzymes. These advances in preparation methods led to the emergence of new generations of nanosystems that extend beyond transport vehicles to diverse applications, from multifunctional drug delivery to imaging, nanocatalysis and protein therapy.
Collapse
Affiliation(s)
- Jonas Kaltbeitzel
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
22
|
Princen K, Marien N, Guedens W, Graulus GJ, Adriaensens P. Hydrogels with Reversible Crosslinks for Improved Localised Stem Cell Retention: A Review. Chembiochem 2023; 24:e202300149. [PMID: 37220343 DOI: 10.1002/cbic.202300149] [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: 02/24/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 05/25/2023]
Abstract
Successful stem cell applications could have a significant impact on the medical field, where many lives are at stake. However, the translation of stem cells to the clinic could be improved by overcoming challenges in stem cell transplantation and in vivo retention at the site of tissue damage. This review aims to showcase the most recent insights into developing hydrogels that can deliver, retain, and accommodate stem cells for tissue repair. Hydrogels can be used for tissue engineering, as their flexibility and water content makes them excellent substitutes for the native extracellular matrix. Moreover, the mechanical properties of hydrogels are highly tuneable, and recognition moieties to control cell behaviour and fate can quickly be introduced. This review covers the parameters necessary for the physicochemical design of adaptable hydrogels, the variety of (bio)materials that can be used in such hydrogels, their application in stem cell delivery and some recently developed chemistries for reversible crosslinking. Implementing physical and dynamic covalent chemistry has resulted in adaptable hydrogels that can mimic the dynamic nature of the extracellular matrix.
Collapse
Affiliation(s)
- Ken Princen
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Neeve Marien
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Geert-Jan Graulus
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| |
Collapse
|
23
|
Attia SA, Truong AT, Phan A, Lee SJ, Abanmai M, Markanovic M, Avila H, Luo H, Ali A, Sreekumar PG, Kannan R, MacKay JA. αB-Crystallin Peptide Fused with Elastin-like Polypeptide: Intracellular Activity in Retinal Pigment Epithelial Cells Challenged with Oxidative Stress. Antioxidants (Basel) 2023; 12:1817. [PMID: 37891896 PMCID: PMC10604459 DOI: 10.3390/antiox12101817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Oxidative stress-induced retinal degeneration is among the main contributing factors of serious ocular pathologies that can lead to irreversible blindness. αB-crystallin (cry) is an abundant component of the visual pathway in the vitreous humor, which modulates protein and cellular homeostasis. Within this protein exists a 20 amino acid fragment (mini-cry) with both chaperone and antiapoptotic activity. This study fuses this mini-cry peptide to two temperature-sensitive elastin-like polypeptides (ELP) with the goal of prolonging its activity in the retina. METHODS The biophysical properties and chaperone activity of cry-ELPs were confirmed by mass spectrometry, cloud-point determination, and dynamic light scattering 'DLS'. For the first time, this work compares a simpler ELP architecture, cry-V96, with a previously reported ELP diblock copolymer, cry-SI. Their relative mechanisms of cellular uptake and antiapoptotic potential were tested using retinal pigment epithelial cells (ARPE-19). Oxidative stress was induced with H2O2 and comparative internalization of both cry-ELPs was made using 2D and 3D culture models. We also explored the role of lysosomal membrane permeabilization by confocal microscopy. RESULTS The results indicated successful ELP fusion, cellular association with both 2D and 3D cultures, which were enhanced by oxidative stress. Both constructs suppressed apoptotic signaling (cleaved caspase-3); however, cry-V96 exhibited greater lysosomal escape. CONCLUSIONS ELP architecture is a critical factor to optimize delivery of therapeutic peptides, such as the anti-apoptotic mini-cry peptide; furthermore, the protection of mini-cry via ELPs is enhanced by lysosomal membrane permeabilization.
Collapse
Affiliation(s)
- Sara Aly Attia
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Anh Tan Truong
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Shin-Jae Lee
- Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA;
| | - Manal Abanmai
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Marinella Markanovic
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Hugo Avila
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Haozhong Luo
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | - Atham Ali
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
| | | | - Ram Kannan
- Doheny Eye Institute, Pasadena, CA 91103, USA; (P.G.S.); (R.K.)
- Stein Eye Institute, Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; (S.A.A.); (A.T.T.); (A.P.); (M.A.); (M.M.); (H.A.); (H.L.); (A.A.)
- Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA;
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
24
|
Tatsubo D, Suyama K, Sakamoto N, Tomohara K, Taniguchi S, Maeda I, Nose T. Determining the Sequence Dependency of Self-Assembly of Elastin-Like Peptides Using Short Peptide Analogues with Shuffled Repetitive Sequences. Biochemistry 2023; 62:2559-2570. [PMID: 37540116 DOI: 10.1021/acs.biochem.3c00146] [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: 08/05/2023]
Abstract
Synthetic elastin-like peptides (ELPs) that possess characteristic tropoelastin-derived hydrophobic repetitive sequences, such as (VPGVG)n, exhibit thermoresponsive reversible self-assembly. Although their thermoresponsive properties have been well-studied, the sequence-dependent and structural requirements for self-assembly remain ambiguous. In particular, it is still unclear whether the amino acid sequences derived from tropoelastin are necessary for self-assembly. In this study, 11 sequence-shuffled ELP analogues based on (FPGVG)5, which is a previously developed short ELP (sELP), were designed to elucidate the sequence-dependent and structural requirements for their self-assembly. Among them, eight shuffled peptides exhibited self-assembling properties, whereas the other three peptides were difficult to dissolve in water. Structural analyses revealed that the structural characteristics of the three insoluble peptides were different from those of their thermoresponsive analogues. Furthermore, the secondary structures of the peptide analogues possessing the self-assembly abilities were different from each other. These results suggest that the potential for self-assembly and water solubility of sELPs depend on the primary structure in each repeated unit. Moreover, several shuffled analogues exhibited more potent self-assembling properties than the original (FPGVG)5, indicating that shorter ELPs can be obtained using their novel motifs as repetitive units. We also observed that the presence of Pro-Gly sequence in the repeating units was advantageous in terms of peptide solubility. Although further analysis will be necessary to elucidate the molecular mechanism underlying the self-assembly of these sELPs, this study provides insights into the relationship between the amino acid sequence and the self-assembling ability of the peptides for developing new sELPs for various applications.
Collapse
Affiliation(s)
- Daiki Tatsubo
- Department of Chemistry, Faculty and Graduate School of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Keitaro Suyama
- Faculty of Arts and Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Naoki Sakamoto
- Department of Chemistry, Faculty and Graduate School of Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Keisuke Tomohara
- Faculty of Arts and Science, Kyushu University, Fukuoka 819-0395, Japan
| | - Suguru Taniguchi
- Department of Physics and Information Technology, Kyushu Institute of Technology, Iizuka 820-8502, Fukuoka, Japan
| | - Iori Maeda
- Department of Physics and Information Technology, Kyushu Institute of Technology, Iizuka 820-8502, Fukuoka, Japan
| | - Takeru Nose
- Department of Chemistry, Faculty and Graduate School of Science, Kyushu University, Fukuoka 819-0395, Japan
- Faculty of Arts and Science, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
25
|
Gowtham P, Arumugam VA, Harini K, Pallavi P, Thirumalai A, Girigoswami K, Girigoswami A. Nanostructured proteins for delivering drugs to diseased tissues. BIOINSPIRED, BIOMIMETIC AND NANOBIOMATERIALS 2023; 12:115-129. [DOI: 10.1680/jbibn.23.00004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
During the last few years, nanostructures based on proteins have been playing a vital role in revolutionizing the nanomedicine era. Since protein nanoparticles are smaller and have a greater surface area, they retain a better capacity to interact with other molecules, resulting in carrying payloads efficiently to diseased tissues. Besides having attractive biocompatibility and biodegradability, protein nanoparticles can also be modified on their surfaces. For the fabrication of these nanostructures, there are several processes involved, including emulsification, desolvation, a combination of complex coacervation and electrospray. This can be achieved by using different proteins such as albumin, gelatin, elastin, gliadin, collagen, legumin and zein, as well as a combination of these proteins. It is possible to functionalize protein nanoparticles by altering their internal and external interfaces so that they can encapsulate drugs, release them in a controlled manner, disassemble them systematically and target tumors. This review highlights the physicochemical properties and engineering of several proteins to nano-dimensions used to deliver drugs to diseased tissues.
Collapse
Affiliation(s)
- Pemula Gowtham
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Vijaya Anand Arumugam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, India
| | - Karthick Harini
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Pragya Pallavi
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Anbazhagan Thirumalai
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, India
| |
Collapse
|
26
|
Depenveiller C, Wong H, Crowet JM, Debelle L, Baud S, Dauchez M, Belloy N. Challenging level of rigid-body approach involving numerical elements (CHLORAINE) applied to repeated elastin peptides. J Struct Biol 2023; 215:107986. [PMID: 37343710 DOI: 10.1016/j.jsb.2023.107986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/10/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023]
Abstract
Elastic proteins and derived biomaterials contain numerous tandemly repeated peptides along their sequences, ranging from a few copies to hundreds. These repetitions are responsible for their biochemical, biological and biomechanical properties. These sequences are considered to be intrinsically disordered, and the variations in their behavior are actually mainly due to their high flexibility and lack of stable secondary structures originating from their unique amino acid sequences. Consequently, the simulation of elastic proteins and large elastomeric biomaterials using classical molecular dynamics is an important challenge. Here, we propose a novel approach that allows the application of the DURABIN protocol to repeated elastin-like peptides (r-ELPs) in a simple way. Four large r-ELPs were studied to evaluate our method, which was developed for simulating extracellular matrix proteins at the mesoscopic scale. After structure clustering applied on molecular dynamic trajectories of constitutive peptides (5-mers and 6-mers), the main conformations were used as starting points to define the corresponding primitives, further used as rigid body fragments in our program. Contributions derived from electrostatic and molecular hydrophobicity potentials were tested to evaluate their influence on the interactions during simple mesoscopic simulations. The CHLORAINE approach, despite the thinner granularity due to the size of the patterns used, was included in the DURABIN protocol and emerges as a promising way to simulate elastic macromolecular systems.
Collapse
Affiliation(s)
- C Depenveiller
- Université de Reims Champagne Ardenne, CNRS, MEDyC UMR 7369, 51097 Reims, France; Université de Picardie Jules Verne, CNRS, GEC UMR 7025, 80039 Amiens, France
| | - H Wong
- Université de Reims Champagne Ardenne, CNRS, MEDyC UMR 7369, 51097 Reims, France; Université de Reims Champagne Ardenne, Plateau de Modélisation Moléculaire Multi-Echelle (P3M), Maison de la simulation de Champagne Ardenne (MaSCA), 51097 Reims, France
| | - J M Crowet
- Université de Reims Champagne Ardenne, CNRS, MEDyC UMR 7369, 51097 Reims, France; Université de Reims Champagne Ardenne, Plateau de Modélisation Moléculaire Multi-Echelle (P3M), Maison de la simulation de Champagne Ardenne (MaSCA), 51097 Reims, France
| | - L Debelle
- Université de Reims Champagne Ardenne, CNRS, MEDyC UMR 7369, 51097 Reims, France
| | - S Baud
- Université de Reims Champagne Ardenne, CNRS, MEDyC UMR 7369, 51097 Reims, France; Université de Reims Champagne Ardenne, Plateau de Modélisation Moléculaire Multi-Echelle (P3M), Maison de la simulation de Champagne Ardenne (MaSCA), 51097 Reims, France
| | - M Dauchez
- Université de Reims Champagne Ardenne, CNRS, MEDyC UMR 7369, 51097 Reims, France; Université de Reims Champagne Ardenne, Plateau de Modélisation Moléculaire Multi-Echelle (P3M), Maison de la simulation de Champagne Ardenne (MaSCA), 51097 Reims, France
| | - N Belloy
- Université de Reims Champagne Ardenne, CNRS, MEDyC UMR 7369, 51097 Reims, France; Université de Reims Champagne Ardenne, Plateau de Modélisation Moléculaire Multi-Echelle (P3M), Maison de la simulation de Champagne Ardenne (MaSCA), 51097 Reims, France.
| |
Collapse
|
27
|
Woo SH, Choi JH, Mo YJ, Lee YI, Jeon WB, Lee YS. Engineered elastin-like polypeptide improves the efficiency of adipose-derived stem cell-mediated cutaneous wound healing in type II diabetes mellitus. Heliyon 2023; 9:e20201. [PMID: 37809635 PMCID: PMC10559957 DOI: 10.1016/j.heliyon.2023.e20201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Impaired cutaneous wound healing is a major complication in patients with diabetes mellitus (DM), leading to increased amputation and mortality rates in affected patients. Adipose-derived stem cells (ASCs) are widely used seed cells for promoted tissue regeneration to improve wound closure under diabetic conditions. However, ASCs-based therapies remain limited due to difficulties in maintaining cell quality during transplantation. To overcome this problem, extracellular matrix mimetic biomaterials have been developed for use in biomedical engineering field, including tissue engineering and regenerative medicine. Herein, a biosynthesized arginine-glycine-aspartate amino acid residues (RGD motif, known as a cell adhesion motif)-containing elastin-like polypeptides (REPs) improved the efficacy of ASCs in enhancing wound closure and skin elasticity in diabetic wounds by promoting the expression of angiogenic growth factors. Therefore, REPs can be used as potential supplements to stem cell-based therapeutic approach to accelerate diabetic wound repair.
Collapse
Affiliation(s)
- Seung-Hwa Woo
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Joon Hyuk Choi
- Division of Biotechnology, DGIST, Daegu, 42988, Republic of Korea
| | - Yun Jeong Mo
- Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988, Republic of Korea
| | - Won Bae Jeon
- Division of Biotechnology, DGIST, Daegu, 42988, Republic of Korea
| | - Young-Sam Lee
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
- Well Aging Research Center, Division of Biotechnology, DGIST, Daegu, 42988, Republic of Korea
| |
Collapse
|
28
|
Kang MJ, Roh KH, Lee JS, Lee JH, Park S, Lim DW. Vascular Endothelial Growth Factor Receptor 1 Targeting Fusion Polypeptides with Stimuli-Responsiveness for Anti-angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37384534 DOI: 10.1021/acsami.3c03989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Genetically engineered fusion polypeptides have been investigated to introduce unique bio-functionality and improve some therapeutic activity for anti-angiogenesis. We report herein that stimuli-responsive, vascular endothelial growth factor receptor 1 (VEGFR1) targeting fusion polypeptides composed of a VEGFR1 (fms-like tyrosine kinase-1 (Flt1)) antagonist, an anti-Flt1 peptide, and a thermally responsive elastin-based polypeptide (EBP) were rationally designed at the genetic level, biosynthesized, and purified by inverse transition cycling to develop potential anti-angiogenic fusion polypeptides to treat neovascular diseases. A series of hydrophilic EBPs with different block lengths were fused with an anti-Flt1 peptide, forming anti-Flt1-EBPs, and the effect of EBP block length on their physicochemical properties was examined. While the anti-Flt1 peptide decreased phase-transition temperatures of anti-Flt1-EBPs, compared with EBP blocks, anti-Flt1-EBPs were soluble under physiological conditions. The anti-Flt1-EBPs dose dependently inhibited the binding of VEGFR1 against vascular endothelial growth factor (VEGF) as well as tube-like network formation of human umbilical vein endothelial cells under VEGF-triggered angiogenesis in vitro because of the specific binding between anti-Flt1-EBPs and VEGFR1. Furthermore, the anti-Flt1-EBPs suppressed laser-induced choroidal neovascularization in a wet age-related macular degeneration mouse model in vivo. Our results indicate that anti-Flt1-EBPs as VEGFR1-targeting fusion polypeptides have great potential for efficacious anti-angiogenesis to treat retinal-, corneal-, and choroidal neovascularization.
Collapse
Affiliation(s)
- Min Jeong Kang
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Kug-Hwan Roh
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Jae Sang Lee
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jae Hee Lee
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - SaeGwang Park
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Dong Woo Lim
- Department of Bionano Engineering and Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
29
|
Li Y, Dautel DR, Gray MA, McKenna ME, Champion JA. Rational design of elastin-like polypeptide fusion proteins to tune self-assembly and properties of protein vesicles. J Mater Chem B 2023. [PMID: 37357544 DOI: 10.1039/d3tb00200d] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Protein vesicles made from bioactive proteins have potential value in drug delivery, biocatalysis, and as artificial cells. As the proteins are produced recombinantly, the ability to precisely tune the protein sequence provides control not possible with polymeric vesicles. The tunability and biocompatibility motivated this work to develop protein vesicles using rationally designed protein building blocks to investigate how protein sequence influences vesicle self-assembly and properties. We have reported an elastin-like polypeptide (ELP) fused to an arginine-rich leucine zipper (ZR) and functional, globular proteins fused to a glutamate-rich leucine zipper (ZE) that self-assemble into protein vesicles when warmed from 4 to 25 °C due to the hydrophobic transition of ELP. Previously, we demonstrated the ability to tune vesicle properties by changing protein and salt concentration, ZE : ZR ratio, and warming rate. However, there is a limit to the properties that can be achieved via assembly conditions. In order to access a wider range of vesicle diameter and stability profiles, this work investigated how modifiying the hydrophobicity and length of the ELP sequence influenced self-assembly and the final properties of protein vesicles using mCherry as a model globular protein. The results showed that both transition temperature and diameter of protein vesicles were inversely correlated to the ELP guest residue hydrophobicity and the number of ELP pentapeptide repeats. Additionally, sequence manipulation enabled assembly of vesicles with properties not accessible by changes to assembly conditions. For example, introduction of tyrosine at 5 guest residue positions in ELP enabled formation of nanoscale vesicles stable at physiological salt concentration. This work yields design guidelines for modifying the ELP sequence to manipulate protein vesicle transition temperature, size and stability to achieve desired properties for particular biofunctional applications.
Collapse
Affiliation(s)
- Yirui Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia, 30332, USA.
- BioEngineering Program, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia, 30332, USA
| | - Dylan R Dautel
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia, 30332, USA.
| | - Mikaela A Gray
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia, 30332, USA.
| | - Michael E McKenna
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia, 30332, USA.
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia, 30332, USA.
- BioEngineering Program, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia, 30332, USA
| |
Collapse
|
30
|
Lee KZ, Jeon J, Jiang B, Subramani SV, Li J, Zhang F. Protein-Based Hydrogels and Their Biomedical Applications. Molecules 2023; 28:4988. [PMID: 37446650 DOI: 10.3390/molecules28134988] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Hydrogels made from proteins are attractive materials for diverse medical applications, as they are biocompatible, biodegradable, and amenable to chemical and biological modifications. Recent advances in protein engineering, synthetic biology, and material science have enabled the fine-tuning of protein sequences, hydrogel structures, and hydrogel mechanical properties, allowing for a broad range of biomedical applications using protein hydrogels. This article reviews recent progresses on protein hydrogels with special focus on those made of microbially produced proteins. We discuss different hydrogel formation strategies and their associated hydrogel properties. We also review various biomedical applications, categorized by the origin of protein sequences. Lastly, current challenges and future opportunities in engineering protein-based hydrogels are discussed. We hope this review will inspire new ideas in material innovation, leading to advanced protein hydrogels with desirable properties for a wide range of biomedical applications.
Collapse
Affiliation(s)
- Kok Zhi Lee
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Juya Jeon
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Bojing Jiang
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Shri Venkatesh Subramani
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Jingyao Li
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| | - Fuzhong Zhang
- Department of Energy, Environmental & Chemical Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
- Institute of Materials Science and Engineering, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
- Division of Biological & Biomedical Sciences, Washington University in St. Louis, One Brookings Drive, Saint Louis, MI 63130, USA
| |
Collapse
|
31
|
Sambani K, Kontomaris SV, Yova D. Atomic Force Microscopy Imaging of Elastin Nanofibers Self-Assembly. MATERIALS (BASEL, SWITZERLAND) 2023; 16:4313. [PMID: 37374496 DOI: 10.3390/ma16124313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
Elastin is an extracellular matrix protein, providing elasticity to the organs, such as skin, blood vessels, lungs and elastic ligaments, presenting self-assembling ability to form elastic fibers. The elastin protein, as a component of elastin fibers, is one of the major proteins found in connective tissue and is responsible for the elasticity of tissues. It provides resilience to the human body, assembled as a continuous mesh of fibers that require to be deformed repetitively and reversibly. Thus, it is of great importance to investigate the development of the nanostructural surface of elastin-based biomaterials. The purpose of this research was to image the self-assembling process of elastin fiber structure under different experimental parameters such as suspension medium, elastin concentration, temperature of stock suspension and time interval after the preparation of the stock suspension. atomic force microscopy (AFM) was applied in order to investigate how different experimental parameters affected fiber development and morphology. The results demonstrated that through altering a number of experimental parameters, it was possible to affect the self-assembly procedure of elastin fibers from nanofibers and the formation of elastin nanostructured mesh consisting of naturally occurring fibers. Further clarification of the contribution of different parameters on fibril formation will enable the design and control of elastin-based nanobiomaterials with predetermined characteristics.
Collapse
Affiliation(s)
- Kyriaki Sambani
- Biomedical Optics and Applied Biophysics Laboratory, Division of Electromagnetics, School of Electrical and Computer Engineering, Electrooptics and Electronic Materials, National Technical University of Athens, 9, Iroon Polytechniou, 15780 Athens, Greece
| | - Stylianos Vasileios Kontomaris
- Faculty of Engineering and Architecture, Metropolitan College, 15125 Athens, Greece
- BioNanoTec Ltd., 2404 Nicosia, Cyprus
| | - Dido Yova
- Biomedical Optics and Applied Biophysics Laboratory, Division of Electromagnetics, School of Electrical and Computer Engineering, Electrooptics and Electronic Materials, National Technical University of Athens, 9, Iroon Polytechniou, 15780 Athens, Greece
| |
Collapse
|
32
|
Goel R, Gulwani D, Upadhyay P, Sarangthem V, Singh TD. Unsung versatility of elastin-like polypeptide inspired spheroid fabrication: A review. Int J Biol Macromol 2023; 234:123664. [PMID: 36791934 DOI: 10.1016/j.ijbiomac.2023.123664] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Lately, 3D cell culture technique has gained a lot of appreciation as a research model. Augmented with technological advancements, the area of 3D cell culture is growing rapidly with a diverse array of scaffolds being tested. This is especially the case for spheroid cultures. The culture of cells as spheroids provides opportunities for unanticipated vision into biological phenomena with its application to drug discovery, metabolic profiling, stem cell research as well as tumor, and disease biology. Spheroid fabrication techniques are broadly categorised into matrix-dependent and matrix-independent techniques. While there is a profusion of spheroid fabrication substrates with substantial biological relevance, an economical, modular, and bio-compatible substrate for high throughput production of spheroids is lacking. In this review, we posit the prospects of elastin-like polypeptides (ELPs) as a broad-spectrum spheroid fabrication platform. Elastin-like polypeptides are nature inspired, size-tunable genetically engineered polymers with wide applicability in various arena of biological considerations, has been employed for spheroid culture with profound utility. The technology offers a cheap, high-throughput, reproducible alternative for spheroid culture with exquisite adaptability. Here, we will brief the applicability of 3D cultures as compared to 2D cultures with spheroids being the focal point of the review. Common approaches to spheroid fabrication are discussed with existential limitations. Finally, the versatility of elastin-like polypeptide inspired substrates for spheroid culture has been discussed.
Collapse
Affiliation(s)
- Ridhima Goel
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Deepak Gulwani
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Priyanka Upadhyay
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijaya Sarangthem
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Thoudam Debraj Singh
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
33
|
Kaur M, Gaba J, Singh K, Bhatia Y, Singh A, Singh N. Recent Advances in Recognition Receptors for Electrochemical Biosensing of Mycotoxins-A Review. BIOSENSORS 2023; 13:391. [PMID: 36979603 PMCID: PMC10046307 DOI: 10.3390/bios13030391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Mycotoxins are naturally occurring toxic secondary metabolites produced by fungi in cereals and foodstuffs during the stages of cultivation and storage. Electrochemical biosensing has emerged as a rapid, efficient, and economical approach for the detection and quantification of mycotoxins in different sample media. An electrochemical biosensor consists of two main units, a recognition receptor and a signal transducer. Natural or artificial antibodies, aptamers, molecularly imprinted polymers (MIP), peptides, and DNAzymes have been extensively employed as selective recognition receptors for the electrochemical biosensing of mycotoxins. This article affords a detailed discussion of the recent advances and future prospects of various types of recognition receptors exploited in the electrochemical biosensing of mycotoxins.
Collapse
Affiliation(s)
- Manpreet Kaur
- Department of Chemistry, Punjab Agricultural University, Ludhiana 141004, India
| | - Jyoti Gaba
- Department of Chemistry, Punjab Agricultural University, Ludhiana 141004, India
| | - Komal Singh
- Department of Chemistry, Punjab Agricultural University, Ludhiana 141004, India
| | - Yashika Bhatia
- Department of Chemistry, Punjab Agricultural University, Ludhiana 141004, India
| | - Anoop Singh
- Department of Chemistry, Indian Institute of Technology, Ropar 140001, India
| | - Narinder Singh
- Department of Chemistry, Indian Institute of Technology, Ropar 140001, India
| |
Collapse
|
34
|
Recent Progress in Proteins-Based Micelles as Drug Delivery Carriers. Polymers (Basel) 2023; 15:polym15040836. [PMID: 36850121 PMCID: PMC9964340 DOI: 10.3390/polym15040836] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Proteins-derived polymeric micelles have gained attention and revolutionized the biomedical field. Proteins are considered a favorable choice for developing micelles because of their biocompatibility, harmlessness, greater blood circulation and solubilization of poorly soluble drugs. They exhibit great potential in drug delivery systems as capable of controlled loading, distribution and function of loaded agents to the targeted sites within the body. Protein micelles successfully cross biological barriers and can be incorporated into various formulation designs employed in biomedical applications. This review emphasizes the recent advances of protein-based polymeric micelles for drug delivery to targeted sites of various diseases. Most studied protein-based micelles such as soy, gelatin, casein and collagen are discussed in detail, and their applications are highlighted. Finally, the future perspectives and forthcoming challenges for protein-based polymeric micelles have been reviewed with anticipated further advances.
Collapse
|
35
|
Hou J, Li N, Zhang W, Zhang W. Exploring the impact of PEGylation on the cell-nanomicelle interactions by AFM-based single-molecule force spectroscopy and force tracing. Acta Biomater 2023; 157:310-320. [PMID: 36535567 DOI: 10.1016/j.actbio.2022.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/15/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
PEGylation has been considered the gold standard method for the modification of various drug delivery systems since the last century. However, the impact of PEGylation on the dynamic interaction between drug carriers and cell membranes has not been quantitatively clarified. Herein, the cellular binding and receptor-mediated endocytosis of a model PEGylated polypeptide nanomicelle were systematically investigated at the single-particle level using AFM-based single-molecule force spectroscopy (SMFS) and force tracing. A self-assembled elastin-like polypeptide (ELP) nanomicelle, which is capable of cross-linking, gastrin-releasing peptide (GRP) modification, and PEGylation was prepared. The cross-linked ELP-based nanomicelles exhibited outstanding stability in a broad temperature range of 4-40 °C, which facilitate the drug loading, as well as our cell-nanomicelle study at the single particle level. The unbinding force between the cross-linked ELP-based nanomicelles and the GRP receptor (GRPR)-containing cell (PC-3) membranes was quantitatively measured by AFM-SMFS. It is found that the PEGylated GRP-displaying nanomicelles exhibit the highest unbinding force, indicating the enhanced specific binding effect of PEGylation. Furthermore, the receptor-mediated endocytosis of the cross-linked ELP-based nanomicelles was monitored with the help of force tracing based on AFM-SMFS. Our results show that PEGylation decreases the endocytic force, duration, and engulfment depth of the PEGylated GRP-displaying nanomicelles, but increases their endocytic velocity, which results from the elimination of non-specific interactions during endocytosis. These observations demonstrate the diverse and complex roles of PEGylation on the interaction of polypeptide nanomicelles to cell membranes and may shed light on the rational design of organic polymer-based drug delivery systems aiming for active and passive targeting strategies. STATEMENT OF SIGNIFICANCE: A self-assembled elastin-like polypeptide (ELP) nanomicelle, which can be easily cross-linked, gastrin-releasing peptide (GRP) modified, and PEGylated, is designed. The AFM-SMFS experiment shows that PEGylation can enhance specific binding of the nanomicelles to the receptors on cell membranes. The force tracing experiment indicates that PEGylation decreases the endocytic force as well as engulfment depth of the nanomicelles through the elimination of non-specific interactions. PEGylation can benefit the drug delivery systems aiming at active targeting, while might not be an ideal modification for drug carriers designed for passive targeting, whose cellular uptake mainly depends on non-specific interactions.
Collapse
Affiliation(s)
- Jue Hou
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Nan Li
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Wei Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China; College of Chemistry, Jilin University, Changchun 130012, PR China.
| | - Wenke Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| |
Collapse
|
36
|
Gallegos A, Wu J. A Molecular Theory of Polypeptide Adsorption at Inorganic Surfaces. J Phys Chem B 2023; 127:794-805. [PMID: 36521053 DOI: 10.1021/acs.jpcb.2c06607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A faithful description of polypeptide adsorption at ionizable surfaces remains a theoretical challenge from a molecular perspective due to the strong coupling of local thermodynamic nonideality and ionizations of both the adsorbate and substrate that are sensitive to the solution condition such as pH, ion valence, and salt concentration. Building upon a recently developed coarse-grained model for natural amino acids in bulk electrolyte solutions, here we report a molecular theory applicable to polypeptide adsorption on ionizable inorganic surfaces over a broad range of inhomogeneous conditions. Our thermodynamic model is able to account for diverse solution effects as well as the amino-acid sequence on polypeptide adsorption and surface association such as hydrogen bonding or bidentate coordination. The theoretical predictions have been validated by extensive comparison with experimental data for the adsorption isotherms of three representative polypeptides at a titanium surface.
Collapse
Affiliation(s)
- Alejandro Gallegos
- Department of Chemical and Environmental Engineering, University of California, Riverside, California92521, United States
| | - Jianzhong Wu
- Department of Chemical and Environmental Engineering, University of California, Riverside, California92521, United States
| |
Collapse
|
37
|
Elastin-like polypeptide-based micelles as a promising platform in nanomedicine. J Control Release 2023; 353:713-726. [PMID: 36526018 DOI: 10.1016/j.jconrel.2022.12.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
New and improved nanomaterials are constantly being developed for biomedical purposes. Nanomaterials based on elastin-like polypeptides (ELPs) have increasingly shown potential over the past two decades. These polymers are artificial proteins of which the design is based on human tropoelastin. Due to this similarity, ELP-based nanomaterials are biodegradable and therefore well suited to drug delivery. The assembly of ELP molecules into nanoparticles spontaneously occurs at temperatures above a transition temperature (Tt). The ELP sequence influences both the Tt and the physicochemical properties of the assembled nanomaterial. Nanoparticles with desired properties can hence be designed by choosing the appropriate sequence. A promising class of ELP nanoparticles are micelles assembled from amphiphilic ELP diblock copolymers. Such micelles are generally uniform and well defined. Furthermore, site-specific attachment of cargo to the hydrophobic block results in micelles with the cargo shielded inside their core, while conjugation to the hydrophilic block causes the cargo to reside in the corona where it is available for interactions. Such control over particle design is one of the main contributing factors for the potential of ELP-based micelles as a drug delivery system. Additionally, the micelles are easily loaded with protein or peptide-based cargo by expressing it as a fusion protein. Small molecule drugs and other cargo types can be either covalently conjugated to ELP domains or physically entrapped inside the micelle core. This review aims to give an overview of ELP-based micelles and their applications in nanomedicine.
Collapse
|
38
|
Garanger E, Lecommandoux S. Emerging opportunities in bioconjugates of Elastin-like polypeptides with synthetic or natural polymers. Adv Drug Deliv Rev 2022; 191:114589. [PMID: 36323382 DOI: 10.1016/j.addr.2022.114589] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/10/2022] [Accepted: 10/24/2022] [Indexed: 01/24/2023]
Abstract
Nature is an everlasting source of inspiration for chemical and polymer scientists seeking to develop ever more innovative materials with greater performances. Natural structural proteins are particularly scrutinized to design biomimetic materials. Often characterized by repeat peptide sequences, that together interact by inter- and intramolecular interactions and form a 3D skeleton, they contribute to the mechanical properties of individual cells, tissues, organs, and whole organisms. (Numata, K. Polymer Journal 2020, 52, 1043-1056) Among them elastin, and its main repeat sequences, have been a source of intense studies for more than 50 years resulting in the specific research field dedicated to elastin-like polypeptides (ELPs). These are currently widely investigated in different applications, namely protein purification, tissue engineering, and drug delivery, and some technologies based on ELPs are currently explored by several start-up companies. In the present review, we have summarized pioneering contributions on ELPs, progress made in their genetic engineering, and understanding of their thermal behavior and self-assembly properties. Considered as intrinsically disordered protein polymers, we have finally focused on the works where ELPs have been conjugated to other synthetic macromolecules as covalent hybrid, statistical, graft, or block copolymers, highlighting the huge opportunities that have still not been explored so far.
Collapse
Affiliation(s)
- Elisabeth Garanger
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, ENSCBP, 16 Avenue Pey-Berland, Pessac F-33600, France.
| | - Sébastien Lecommandoux
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, ENSCBP, 16 Avenue Pey-Berland, Pessac F-33600, France.
| |
Collapse
|
39
|
Yu B, Wang X, Ding L, Han M, Guo Y. Hydrophilic Natural Polylysine as Drug Nanocarrier for Preparation of Helical Delivery System. Pharmaceutics 2022; 14:pharmaceutics14112512. [PMID: 36432704 PMCID: PMC9696163 DOI: 10.3390/pharmaceutics14112512] [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: 09/19/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Polypeptide materials have clear secondary structure and biodegradability, which can be further modified and functionalized, so that they can be employed as therapeutic agents in clinical applications. PEGylation of polylysine (PEG-PLL) is a kind of safe and effective nanocarrier that is utilized for gene and drug delivery. However, PEG-PLL needs to be produced through chemical synthesis, which is expensive and difficult to obtain. We hope to simplify the nanocarrier and use hydrophilic natural polylysine (PLL) to develop a high-efficacy delivery system. To evaluate the possibility of PLL as nanocarriers, methotrexate (MTX) is selected as a model drug and PEG-PLL is utilized as control nanocarriers. The experimental results showed that PLL is an ideal polypeptide to prepare MTX-loaded PLL nanoparticles (PLL/MTX NPs). Compared with PEG-PLL as nanocarriers, PLL/MTX NPs showed higher drug-loading content (58.9%) and smaller particle sizes (113.7 nm). Moreover, the shape of PLL/MTX NPs was a unique helical nanorod. The PLL/MTX NPs had good storage stability, media stability, and sustained release effect. Animal research demonstrated that PLL/MTX NPs could improve the anti-tumor activity of MTX, the antitumor efficacy is enhanced 1.9-fold and 1.2-fold compared with MTX injection and PEG-PLL/MTX NPs, respectively. To sum up, natural polymer PLL is an ideal nano drug delivery carrier which has potential clinical applications.
Collapse
Affiliation(s)
- Bo Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Lijuan Ding
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
- Correspondence:
| |
Collapse
|
40
|
Recombinant protein polymers as carriers of chemotherapeutic agents. Adv Drug Deliv Rev 2022; 190:114544. [PMID: 36176240 DOI: 10.1016/j.addr.2022.114544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/02/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
Chemotherapy is the standard of care for the treatment of cancer and infectious diseases. However, its use is associated with severe toxicity and resistance arising mainly due to non-specificity, resulting in disease progression. The advancement in recombinant technology has led to the synthesis of genetically engineered protein polymers like Elastin-like polypeptide (ELP), Silk-like polypeptide (SLP), hybrid protein polymers with specific sequences to impart precisely controlled properties and to target proteins that have provided satisfactory preclinical outcomes. Such protein polymers have been exploited for the formulation and delivery of chemotherapeutics for biomedical applications. The use of such polymers has not only solved the limitation of conventional chemotherapy but has also improved the therapeutic index of typical drug delivery systems. This review, therefore, summarizes the development of such advanced recombinant protein polymers designed to deliver chemotherapeutics and also discusses the key challenges associated with their current usage and their application in the future.
Collapse
|
41
|
Gueta O, Amiram M. Expanding the chemical repertoire of protein-based polymers for drug-delivery applications. Adv Drug Deliv Rev 2022; 190:114460. [PMID: 36030987 DOI: 10.1016/j.addr.2022.114460] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/12/2022] [Indexed: 01/24/2023]
Abstract
Expanding the chemical repertoire of natural and artificial protein-based polymers (PBPs) can enable the production of sequence-defined, yet chemically diverse, biopolymers with customized or new properties that cannot be accessed in PBPs composed of only natural amino acids. Various approaches can enable the expansion of the chemical repertoire of PBPs, including chemical and enzymatic treatments or the incorporation of unnatural amino acids. These techniques are employed to install a wide variety of chemical groups-such as bio-orthogonally reactive, cross-linkable, post-translation modifications, and environmentally responsive groups-which, in turn, can facilitate the design of customized PBP-based drug-delivery systems with modified, fine-tuned, or entirely new properties and functions. Here, we detail the existing and emerging technologies for expanding the chemical repertoire of PBPs and review several chemical groups that either demonstrate or are anticipated to show potential in the design of PBP-based drug delivery systems. Finally, we provide our perspective on the remaining challenges and future directions in this field.
Collapse
Affiliation(s)
- Osher Gueta
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel
| | - Miriam Amiram
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel.
| |
Collapse
|
42
|
Gong L, Yang Z, Zhang F, Gao W. Cytokine conjugates to elastin-like polypeptides. Adv Drug Deliv Rev 2022; 190:114541. [PMID: 36126792 DOI: 10.1016/j.addr.2022.114541] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/25/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
Cytokines are a group of pleiotropic proteins which are crucial for various biological processes and useful as therapeutics. However, they usually suffer from the poor stability, extreme short circulation half-life, difficulty in high-yield and large-scale production and side effects, which greatly restricts their applications. Over the past decades, conjugation of cytokines with elastin-like polypeptides (ELPs), a type of promising biomaterials, have showed great potential in solving these challenges due to ELP's thermal responsiveness, excellent biocompatibility and biodegradability, non-immunogenicity, and ease of design and control at the genetic level. This review presents recent progress in the design and production of a variety of ELP conjugated cytokines for extended circulation, enhanced stability, increased soluble protein expression, simplified purification, improved drug delivery, and controlled release. Notably, the unique thermoresponsive properties of cytokine-ELP conjugates make it possible to self-assemble into micelles with drastically extended circulatory half-life for targeted delivery or to in situ form drug depots for topical administration and controlled release. The challenges and issues in the emerging field are further discussed and the future directions are pointed out at the end of this review.
Collapse
Affiliation(s)
- Like Gong
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China; Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing 100081, China; Biomedical Engineering Department, Peking University, Beijing 100191, China; Peking University International Cancer Institute, Beijing 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Zhaoying Yang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China; Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing 100081, China; Biomedical Engineering Department, Peking University, Beijing 100191, China; Peking University International Cancer Institute, Beijing 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Fan Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China; Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing 100081, China; Biomedical Engineering Department, Peking University, Beijing 100191, China; Peking University International Cancer Institute, Beijing 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China
| | - Weiping Gao
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China; Department of Geriatric Dentistry, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing 100081, China; Biomedical Engineering Department, Peking University, Beijing 100191, China; Peking University International Cancer Institute, Beijing 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing 100191, China.
| |
Collapse
|
43
|
Chen LL, Xu YC, Yang Y, Li N, Zou HX, Wen HH, Yan X. Prediction of peptide-induced silica formation under a wide pH range by molecular descriptors. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.130030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
44
|
Li Y, Champion JA. Self-assembling nanocarriers from engineered proteins: Design, functionalization, and application for drug delivery. Adv Drug Deliv Rev 2022; 189:114462. [PMID: 35934126 DOI: 10.1016/j.addr.2022.114462] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 07/15/2022] [Indexed: 01/24/2023]
Abstract
Self-assembling proteins are valuable building blocks for constructing drug nanocarriers due to their self-assembly behavior, monodispersity, biocompatibility, and biodegradability. Genetic and chemical modifications allow for modular design of protein nanocarriers with effective drug encapsulation, targetability, stimuli responsiveness, and in vivo half-life. Protein nanocarriers have been developed to deliver various therapeutic molecules including small molecules, proteins, and nucleic acids with proven in vitro and in vivo efficacy. This article reviews recent advances in protein nanocarriers that are not derived from natural protein nanostructures, such as protein cages or virus like particles. The protein nanocarriers described here are self-assembled from rationally or de novo designed recombinant proteins, as well as recombinant proteins complexed with other biomolecules, presenting properties that are unique from those of natural protein carriers. Design, functionalization, and therapeutic application of protein nanocarriers will be discussed.
Collapse
Affiliation(s)
- Yirui Li
- BioEngineering Program, Georgia Institute of Technology, United States
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, United States; BioEngineering Program, Georgia Institute of Technology, United States.
| |
Collapse
|
45
|
Levêque M, Xiao Y, Durand L, Massé L, Garanger E, Lecommandoux S. Aqueous synthesis and self-assembly of bioactive and thermo-responsive HA- b-ELP bioconjugates. Biomater Sci 2022; 10:6365-6376. [PMID: 36168976 DOI: 10.1039/d2bm01149b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The design of synthetic (bio)macromolecules that combine biocompatibility, self-assembly and bioactivity properties at the molecular level is an intense field of research for biomedical applications such as (nano)medicine. In this contribution, we have designed and synthesized a library of bioactive and thermo-responsive bioconjugates from elastin-like polypeptides (ELPs) and hyaluronic acid (HA) in order to access bioactive self-assembled nanoparticles. These were prepared by a simple synthetic and purification strategy, compatible with the requirements for biological applications and industrial scale-up. A series of 9 HA-b-ELP bioconjugates with different compositions and block lengths was synthesized under aqueous conditions by strain-promoted azide-alkyne cycloaddition (SPAAC), avoiding the use of catalysts, co-reactants and organic solvents, and isolated by a simple centrifugation step. An extensive physico-chemical study was then performed on the whole library of bioconjugates in an attempt to establish structure-property relationships. In particular, the determination of the critical conditions for thermally driven self-assembly was carried out upon temperature (CMT) and concentration (CMC) gradients, leading to a phase diagram for each of these bioconjugates. These parameters and the size of nanoparticles were found to depend on the chemical composition of the bioconjugates, namely on the respective size of individual blocks. Understanding the mechanism underlying this dependency is a real asset for designing more effective experiments: with key criteria defined (e.g. concentration, temperature, salinity, and biological target), the composition of the best candidates can be rationalized. In particular, four of the bioconjugates (HA4.6k-ELPn80 or n100 and HA24k-ELPn80 or n100) were found to self-assemble into well-defined spherical core-shell nanoparticles, with a negative surface charge due to the HA block exposed at the surface, a hydrodynamic diameter between 40 and 200 nm under physiological conditions and a good stability over time at 37 °C. We therefore propose here a versatile and simple design of smart, controllable, and bioactive nanoparticles that present different behaviors depending on the diblocks' composition.
Collapse
Affiliation(s)
- Manon Levêque
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Ye Xiao
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Laura Durand
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Louise Massé
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Elisabeth Garanger
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | | |
Collapse
|
46
|
Wang K, Zheng M, Askew C, Zhang X, Li C, Han Z. Elastin‐Like Polypeptides Facilitate Adeno‐Associated Virus Transduction in the Presence of Pre‐Existing Neutralizing Antibodies. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Kai Wang
- Department of Ophthalmology The University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Min Zheng
- Department of Ophthalmology The University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Charles Askew
- Gene Therapy Center The University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Xintao Zhang
- Gene Therapy Center The University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Chengwen Li
- Gene Therapy Center The University of North Carolina at Chapel Hill Chapel Hill NC USA
- Department of Pediatrics The University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Zongchao Han
- Department of Ophthalmology The University of North Carolina at Chapel Hill Chapel Hill NC USA
- Pharmacoengineering & Molecular Pharmaceutics UNC Eshelman School of Pharmacy Chapel Hill NC USA
| |
Collapse
|
47
|
Alvisi N, Zheng C, Lokker M, Boekestein V, de Haas R, Albada B, de Vries R. Design of Polypeptides Self-Assembling into Antifouling Coatings: Exploiting Multivalency. Biomacromolecules 2022; 23:3507-3516. [PMID: 35952369 PMCID: PMC9472226 DOI: 10.1021/acs.biomac.2c00170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We propose to exploit multivalent binding of solid-binding peptides (SBPs) for the physical attachment of antifouling polypeptide brushes on solid surfaces. Using a silica-binding peptide as a model SBP, we find that both tandem-repeated SBPs and SBPs repeated in branched architectures implemented via a multimerization domain work very well to improve the binding strength of polypeptide brushes, as compared to earlier designs with a single SBP. At the same time, for many of the designed sequences, either the solubility or the yield of recombinant production is low. For a single design, with the domain structure B-M-E, both solubility and yield of recombinant production were high. In this design, B is a silica-binding peptide, M is a highly thermostable, de novo-designed trimerization domain, and E is a hydrophilic elastin-like polypeptide. We show that the B-M-E triblock polypeptide rapidly assembles into highly stable polypeptide brushes on silica surfaces, with excellent antifouling properties against high concentrations of serum albumin. Given that SBPs attaching to a wide range of materials have been identified, the B-M-E triblock design provides a template for the development of polypeptides for coating many other materials such as metals or plastics.
Collapse
Affiliation(s)
- Nicolò Alvisi
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Chuanbao Zheng
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Meike Lokker
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Victor Boekestein
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Robbert de Haas
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Bauke Albada
- Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Renko de Vries
- Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| |
Collapse
|
48
|
Choi J, Heo T, Choi H, Choi S, Won J. Co‐assembly
behavior of oppositely charged thermoresponsive elastin‐like polypeptide block copolymers. J Appl Polym Sci 2022. [DOI: 10.1002/app.52906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jeong‐Wan Choi
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Tae‐Young Heo
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Heelak Choi
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Soo‐Hyung Choi
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| | - Jong‐In Won
- Department of Chemical Engineering Hongik University Seoul Republic of Korea
| |
Collapse
|
49
|
Zhang Y, Gao S, Qi X, Zhu S, Xu S, Liang Y, Kong F, Yang S, Wang R, Wang Y, An Y. Novel biocatalytic strategy of levan: His-ELP-intein-tagged protein purification and biomimetic mineralization. Carbohydr Polym 2022; 288:119398. [DOI: 10.1016/j.carbpol.2022.119398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/08/2022] [Accepted: 03/19/2022] [Indexed: 01/13/2023]
|
50
|
Haas S, Desombre M, Kirschhöfer F, Huber MC, Schiller SM, Hubbuch J. Purification of a Hydrophobic Elastin-Like Protein Toward Scale-Suitable Production of Biomaterials. Front Bioeng Biotechnol 2022; 10:878838. [PMID: 35814018 PMCID: PMC9257828 DOI: 10.3389/fbioe.2022.878838] [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: 02/18/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Elastin-like proteins (ELPs) are polypeptides with potential applications as renewable bio-based high-performance polymers, which undergo a stimulus-responsive reversible phase transition. The ELP investigated in this manuscript—ELP[V2Y-45]—promises fascinating mechanical properties in biomaterial applications. Purification process scalability and purification performance are important factors for the evaluation of potential industrial-scale production of ELPs. Salt-induced precipitation, inverse transition cycling (ITC), and immobilized metal ion affinity chromatography (IMAC) were assessed as purification protocols for a polyhistidine-tagged hydrophobic ELP showing low-temperature transition behavior. IMAC achieved a purity of 86% and the lowest nucleic acid contamination of all processes. Metal ion leakage did not propagate chemical modifications and could be successfully removed through size-exclusion chromatography. The simplest approach using a high-salt precipitation resulted in a 60% higher target molecule yield compared to both other approaches, with the drawback of a lower purity of 60% and higher nucleic acid contamination. An additional ITC purification led to the highest purity of 88% and high nucleic acid removal. However, expensive temperature-dependent centrifugation steps are required and aggregation effects even at low temperatures have to be considered for the investigated ELP. Therefore, ITC and IMAC are promising downstream processes for biomedical applications with scale-dependent economical costs to be considered, while salt-induced precipitation may be a fast and simple alternative for large-scale bio-based polymer production.
Collapse
Affiliation(s)
- Sandra Haas
- Institute of Process Engineering in Life Sciences, Section IV: Molecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Monika Desombre
- Institute of Process Engineering in Life Sciences, Section IV: Molecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Frank Kirschhöfer
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Matthias C. Huber
- Center for Biosystems Analysis, Albert‐Ludwigs‐University Freiburg, Freiburg, Germany
- Cluster of Excellence livMatS @ FIT, Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Stefan M. Schiller
- Center for Biosystems Analysis, Albert‐Ludwigs‐University Freiburg, Freiburg, Germany
- Cluster of Excellence livMatS @ FIT, Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Jürgen Hubbuch
- Institute of Process Engineering in Life Sciences, Section IV: Molecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
- *Correspondence: Jürgen Hubbuch,
| |
Collapse
|