1
|
Narayan OP, Dong J, Huang M, Chen L, Liu L, Nguyen V, Dozic AV, Liu X, Wang H, Yin Q, Tang X, Guan J. Reversible light-responsive protein hydrogel for on-demand cell encapsulation and release. Acta Biomater 2025:S1742-7061(25)00020-0. [PMID: 39800098 DOI: 10.1016/j.actbio.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
The design of biomaterials that can reconfigure on-demand in response to external stimuli is an emerging area in materials research. However, achieving reversible assembly of protein-based biomaterials by light input remains a major challenge. Here, we present the engineering of a new protein material that is capable of switching between liquid and solid state reversibly, controlled by lights of different wavelengths. The materials are created by incorporating a light-responsive mutant Dronpa protein domain into the backbone of Elastin-Like Proteins (termed DELPs). We show that the DELP material can respond to light and undergo multiple cycles of switching between hydrogel and solution, outperforming the conventional irreversible materials. Additionally, the material is biocompatible with long-term cell proliferation in both adherent and suspension cells. Building on the reversible assembly of the material, we demonstrate efficient cell encapsulation and release upon light triggers. The design principle of incorporating a light-responsive protein element into a structural protein matrix, as demonstrated in this work enables, a broad range of other applications that require adaptive materials to intelligently interface with dynamic biological systems and environments. STATEMENT OF SIGNIFICANCE: This work generates a new class of "smart" biomaterials that uniquely switches between liquid and gel states in response to light input. Light input can be precisely delivered in space and time, highly tunable through wavelengths, intensities, and durations of light exposure. In prior research, light-responsive biomaterials are mostly irreversible, limiting their use to only uni-directional applications and the materials cannot be re-used. In contrast, this material robustly displays reversible switching between liquid and gel using a light-responsive crosslinker. Furthermore, the material is biocompatible, programmable, and suitable for broad applications including but not limited to cell encapsulation, controlled release, tissue engineering, and cell/tissue mechanobiology.
Collapse
Affiliation(s)
- Om Prakash Narayan
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jiawei Dong
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Liqiang Chen
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Lu Liu
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Vivian Nguyen
- Department of Biology, University of Florida, Gainesville, FL 32611, USA
| | - Abdul Vehab Dozic
- Department of Physics, University of Florida, Gainesville, FL 32611, USA
| | - Xiangping Liu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Huiliang Wang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Qian Yin
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA.
| | - Juan Guan
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
2
|
Zhang S, Gong L, Sun Y, Zhang F, Gao W. An ultra-long-acting L-asparaginase synergizes with an immune checkpoint inhibitor in starvation-immunotherapy of metastatic solid tumors. Biomaterials 2025; 312:122740. [PMID: 39096839 DOI: 10.1016/j.biomaterials.2024.122740] [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: 12/31/2023] [Revised: 07/05/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
Metastasis stands as the primary contributor to mortality associated with tumors. Chemotherapy and immunotherapy are frequently utilized in the management of metastatic solid tumors. Nevertheless, these therapeutic modalities are linked to serious adverse effects and limited effectiveness in preventing metastasis. Here, we report a novel therapeutic strategy named starvation-immunotherapy, wherein an immune checkpoint inhibitor is combined with an ultra-long-acting L-asparaginase that is a fusion protein comprising L-asparaginase (ASNase) and an elastin-like polypeptide (ELP), termed ASNase-ELP. ASNase-ELP's thermosensitivity enables it to generate an in-situ depot following an intratumoral injection, yielding increased dose tolerance, improved pharmacokinetics, sustained release, optimized biodistribution, and augmented tumor retention compared to free ASNase. As a result, in murine models of oral cancer, melanoma, and cervical cancer, the antitumor efficacy of ASNase-ELP by selectively and sustainably depleting L-asparagine essential for tumor cell survival was substantially superior to that of ASNase or Cisplatin, a first-line anti-solid tumor medicine, without any observable adverse effects. Furthermore, the combination of ASNase-ELP and an immune checkpoint inhibitor was more effective than either therapy alone in impeding melanoma metastasis. Overall, the synergistic strategy of starvation-immunotherapy holds excellent promise in reshaping the therapeutic landscape of refractory metastatic tumors and offering a new alternative for next-generation oncology treatments.
Collapse
Affiliation(s)
- Sanke Zhang
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China
| | - Like Gong
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China
| | - Yuanzi Sun
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, 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
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, 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
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China; Peking University International Cancer Institute, Beijing, 100191, China; Peking University-Yunnan Baiyao International Medical Research Center, Beijing, 100191, China; Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China.
| |
Collapse
|
3
|
Zhang L, Li X, He Q, Chen M, Zhou M, Guo J, Li Y, Tu Z. Elastin-like polypeptide-functionalized nanobody for column-free immunoaffinity purification of aflatoxin B 1. Anal Bioanal Chem 2024; 416:6199-6208. [PMID: 39264463 DOI: 10.1007/s00216-024-05498-0] [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: 03/25/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024]
Abstract
A column-free immunoaffinity purification (CFIP) technique for sample preparation of aflatoxin B1 (AFB1) was developed using an AFB1-specific nanobody (named G8) and an elastin-like polypeptide (ELP). The reversible phase transition between liquid and solid in response to temperature changes was exhibited by the ELP which was derived from human elastin. The G8 was tagged with ELPs of various lengths (20, 40, 60, and 80 repeat units) at the C-terminus using recursive directional ligation (RDL). Coding sequences were then subcloned into pET30a at the multiple cloning sites. Bioactive recombinant proteins were produced by expressing them as inclusion bodies in Escherichia coli BL21 (DE3), then dissolved and refolded. Analysis by indirect competitive enzyme-linked immunosorbent assay (icELISA) and transition temperature (Tt) measurement confirmed that the refolded G8-ELPs preserved the ability to recognize AFB1 as well as phase transition when the temperature rose above Tt. To establish the optimal conditions for cleaning AFB1, the effects of various parameters on recovery were investigated. The recovery in ELISA tests was 95 ± 3.67% under the optimized CFIP workflow. Furthermore, the CFIP-prepared samples were applied for high-performance liquid chromatography (HPLC) detection. The recovery in the CFIP-HPLC test ranged from 54 ± 1.86% to 98 ± 3.58% for maize, rice, soy sauce, and vegetable oil samples. To the best of our knowledge, this is the first report combining the function of both nanobody and ELP to develop a cleanup technique for small molecules in a complex matrix. The CFIP for the sample pretreatment was easy to use and inexpensive. In contrast to conventional immunosensitivity materials, the reagent utilized in the CFIP was entirely biosynthesized without any chemical coupling reactions. This suggests that the nanobody-ELP may serve as a useful dual-functional reagent for the development of sample cleaning or purification methods.
Collapse
Affiliation(s)
- Leping Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Xiaojiang Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Qinghua He
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- Jiangxi-OAI Joint Research Institution, Nanchang University, Nanchang, 330047, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Mengna Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Mengmeng Zhou
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
- College of Food Science, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China
| | - Jiebiao Guo
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, 512005, China
| | - Yanping Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
- Jiangxi-OAI Joint Research Institution, Nanchang University, Nanchang, 330047, China.
| | - Zhui Tu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
- Jiangxi Province Key Laboratory of Modern Analytical Sciences, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
4
|
Kadam S, Ali A, Pospiech M, Onyemaechi S, Meng Y, Dhuri K, MacKay JA, Alachkar H. Enhanced T cell activation and cytotoxicity against AML via targeted anti-CD99 nanoparticle treatment. Biomed Pharmacother 2024; 179:117265. [PMID: 39167846 DOI: 10.1016/j.biopha.2024.117265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
CD99 is a transmembrane protein overexpressed in Acute Myeloid Leukemia (AML), presenting a potential novel therapeutic target. Our group has previously developed anti-CD99-A192 (α-CD99-A192), comprising of single chain variable fragment (scFv) and elastin-like polypeptides (ELPs), and reported promising anti-leukemic activity in AML preclinical models. Treatment with α-CD99-A192 induced apoptosis in AML cell lines and prolonged survival in AML xenograft models. Considering CD99's expression and role in T cell activation, in the current study, we propose that α-CD99-A192 plays a dual function, i.e., targeting leukemic cells and activating T cells. This manuscript reports the effects of α-CD99-A192 on T cells in the context of AML. α-CD99-A192 treatment enhances T cell proliferation and activation and increases the release of pro-inflammatory cytokines along with increased aggregation of T cells, which culminates in heightened cytotoxicity against leukemic cells. Altogether, these findings suggest α-CD99-A192 enhances T cell activation and cytotoxic potential consistent with dual mechanisms of action for α-CD99-A192.
Collapse
Affiliation(s)
- Shephali Kadam
- 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 Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Atham Ali
- Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Mateusz Pospiech
- Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Sandra Onyemaechi
- Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Yiting Meng
- Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Kanaka Dhuri
- 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 Clinical Pharmacy, 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; 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 90033, USA; USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Houda Alachkar
- Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
5
|
Giraldo-Castaño MC, Littlejohn KA, Avecilla ARC, Barrera-Villamizar N, Quiroz FG. Programmability and biomedical utility of intrinsically-disordered protein polymers. Adv Drug Deliv Rev 2024; 212:115418. [PMID: 39094909 PMCID: PMC11389844 DOI: 10.1016/j.addr.2024.115418] [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: 04/20/2024] [Revised: 07/03/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Intrinsically disordered proteins (IDPs) exhibit molecular-level conformational dynamics that are functionally harnessed across a wide range of fascinating biological phenomena. The low sequence complexity of IDPs has led to the design and development of intrinsically-disordered protein polymers (IDPPs), a class of engineered repeat IDPs with stimuli-responsive properties. The perfect repetitive architecture of IDPPs allows for repeat-level encoding of tunable protein functionality. Designer IDPPs can be modeled on endogenous IDPs or engineered de novo as protein polymers with dual biophysical and biological functionality. Their properties can be rationally tailored to access enigmatic IDP biology and to create programmable smart biomaterials. With the goal of inspiring the bioengineering of multifunctional IDP-based materials, here we synthesize recent multidisciplinary progress in programming and exploiting the bio-functionality of IDPPs and IDPP-containing proteins. Collectively, expanding beyond the traditional sequence space of extracellular IDPs, emergent sequence-level control of IDPP functionality is fueling the bioengineering of self-assembling biomaterials, advanced drug delivery systems, tissue scaffolds, and biomolecular condensates -genetically encoded organelle-like structures. Looking forward, we emphasize open challenges and emerging opportunities, arguing that the intracellular behaviors of IDPPs represent a rich space for biomedical discovery and innovation. Combined with the intense focus on IDP biology, the growing landscape of IDPPs and their biomedical applications set the stage for the accelerated engineering of high-value biotechnologies and biomaterials.
Collapse
Affiliation(s)
- Maria Camila Giraldo-Castaño
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kai A Littlejohn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Alexa Regina Chua Avecilla
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Natalia Barrera-Villamizar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Felipe Garcia Quiroz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
6
|
Li D, Wang Y, Zhu S, Hu X, Liang R. Recombinant fibrous protein biomaterials meet skin tissue engineering. Front Bioeng Biotechnol 2024; 12:1411550. [PMID: 39205856 PMCID: PMC11349559 DOI: 10.3389/fbioe.2024.1411550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Natural biomaterials, particularly fibrous proteins, are extensively utilized in skin tissue engineering. However, their application is impeded by batch-to-batch variance, limited chemical or physical versatility, and environmental concerns. Recent advancements in gene editing and fermentation technology have catalyzed the emergence of recombinant fibrous protein biomaterials, which are gaining traction in skin tissue engineering. The modular and highly customizable nature of recombinant synthesis enables precise control over biomaterial design, facilitating the incorporation of multiple functional motifs. Additionally, recombinant synthesis allows for a transition from animal-derived sources to microbial sources, thereby reducing endotoxin content and rendering recombinant fibrous protein biomaterials more amenable to scalable production and clinical use. In this review, we provide an overview of prevalent recombinant fibrous protein biomaterials (collagens, elastin, silk proteins and their chimeric derivatives) used in skin tissue engineering (STE) and compare them with their animal-derived counterparts. Furthermore, we discuss their applications in STE, along with the associated challenges and future prospects.
Collapse
Affiliation(s)
- Dipeng Li
- Hangzhou Ninth People’s Hospital, Hangzhou, China
| | - Yirong Wang
- Hangzhou Singclean Medical Products Co., Ltd., Hangzhou, China
| | - Shan Zhu
- Hangzhou Singclean Medical Products Co., Ltd., Hangzhou, China
| | - Xuezhong Hu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, China
| | - Renjie Liang
- Hangzhou Ninth People’s Hospital, Hangzhou, China
- Hangzhou Singclean Medical Products Co., Ltd., Hangzhou, China
- School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
7
|
Wirtz BM, Yun AG, Wick C, Gao XJ, Mai DJ. Protease-Driven Phase Separation of Elastin-Like Polypeptides. Biomacromolecules 2024; 25:4898-4904. [PMID: 38980747 DOI: 10.1021/acs.biomac.4c00346] [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: 07/11/2024]
Abstract
Elastin-like polypeptides (ELPs) are a promising material platform for engineering stimuli-responsive biomaterials, as ELPs undergo phase separation above a tunable transition temperature. ELPs with phase behavior that is isothermally regulated by biological stimuli remain attractive for applications in biological systems. Herein, we report protease-driven phase separation of ELPs. Protease-responsive "cleavable" ELPs comprise a hydrophobic ELP block connected to a hydrophilic ELP block by a protease cleavage site linker. The hydrophilic ELP block acts as a solubility tag for the hydrophobic ELP block, creating a temperature window in which the cleavable ELP reactant is soluble and the proteolytically generated hydrophobic ELP block is insoluble. Within this temperature window, isothermal, protease-driven phase separation occurs when a critical concentration of hydrophobic cleavage product accumulates. Furthermore, protease-driven phase separation is generalizable to four compatible protease-cleavable ELP pairings. This work presents exciting opportunities to regulate ELP phase behavior in biological systems using proteases.
Collapse
Affiliation(s)
- Brendan M Wirtz
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Allison G Yun
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Chloe Wick
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Xiaojing J Gao
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Danielle J Mai
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
8
|
Tang B, Xie X, Lu J, Huang W, Yang J, Tian J, Lei L. Designing biomaterials for the treatment of autoimmune diseases. APPLIED MATERIALS TODAY 2024; 39:102278. [DOI: 10.1016/j.apmt.2024.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
9
|
Patkar SS, Wang B, Mosquera AM, Kiick KL. Genetically Fusing Order-Promoting and Thermoresponsive Building Blocks to Design Hybrid Biomaterials. Chemistry 2024; 30:e202400582. [PMID: 38501912 PMCID: PMC11661552 DOI: 10.1002/chem.202400582] [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: 02/13/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 03/20/2024]
Abstract
The unique biophysical and biochemical properties of intrinsically disordered proteins (IDPs) and their recombinant derivatives, intrinsically disordered protein polymers (IDPPs) offer opportunities for producing multistimuli-responsive materials; their sequence-encoded disorder and tendency for phase separation facilitate the development of multifunctional materials. This review highlights the strategies for enhancing the structural diversity of elastin-like polypeptides (ELPs) and resilin-like polypeptides (RLPs), and their self-assembled structures via genetic fusion to ordered motifs such as helical or beta sheet domains. In particular, this review describes approaches that harness the synergistic interplay between order-promoting and thermoresponsive building blocks to design hybrid biomaterials, resulting in well-structured, stimuli-responsive supramolecular materials ordered on the nanoscale.
Collapse
Affiliation(s)
- Sai S Patkar
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, 19716, United States
- Eli Lilly and Company, 450 Kendall Street, Cambridge, MA, 02142, United States
| | - Bin Wang
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, 19716, United States
| | - Ana Maria Mosquera
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, 19716, United States
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, 19716, United States
| |
Collapse
|
10
|
Zhou Z, Wang S, Feng T, Zhang P, Fan H, Zou J, Huang K. Nanoassembly-Mediated Exendin-4 Derivatives to Decrease Renal Retention. ACS OMEGA 2024; 9:18757-18765. [PMID: 38708210 PMCID: PMC11064033 DOI: 10.1021/acsomega.3c04644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 05/07/2024]
Abstract
An Exendin-4 analogue that was conjugated with 68Ga exhibited an excellent diagnostic effect on insulinoma in clinical practice. On account of its low molecular weight and short hydration radius, 68Ga-Exendin-4 showed high accumulation in kidney tissues. Nanoparticle-mediated strategies have attracted much attention due to polyvalent properties and the size amplification effect. In this study, Exendin-4 derivatives of radionuclide nanodevices were developed and evaluated. The Exendin-4 derivatives consisting of a ternary block recombinant protein were purified by an inverse transition cycle (ITC) and allowed to self-assemble into a nanodevice under physiological conditions. Our results showed that the nanoassemblies of Exendin-4 derivatives formed homogeneous spherical nanoparticles, exhibited outstanding affinity for insulinoma cells, and could be deposited in insulinoma tissues in vivo. The nanoassembly-mediated Exendin-4 derivatives showed fivefold reduced renal retention and exhibited an outstanding tumor-suppression effect.
Collapse
Affiliation(s)
- Zhengwei Zhou
- Department
of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing 210006, P. R. China
- School
of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, P. R. China
| | - Shuai Wang
- Department
of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing 210006, P. R. China
- School
of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, P. R. China
| | - Tianling Feng
- Department
of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing 210006, P. R. China
- School
of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, P. R. China
| | - Pengjun Zhang
- Department
of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing 210006, P. R. China
| | - Hongwei Fan
- Department
of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing 210006, P. R. China
- School
of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, P. R. China
| | - Jianjun Zou
- Department
of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing 210006, P. R. China
- School
of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, P. R. China
| | - Kaizong Huang
- Department
of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing 210006, P. R. China
| |
Collapse
|
11
|
Gil-Garcia M, Benítez-Mateos AI, Papp M, Stoffel F, Morelli C, Normak K, Makasewicz K, Faltova L, Paradisi F, Arosio P. Local environment in biomolecular condensates modulates enzymatic activity across length scales. Nat Commun 2024; 15:3322. [PMID: 38637545 PMCID: PMC11026464 DOI: 10.1038/s41467-024-47435-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 03/28/2024] [Indexed: 04/20/2024] Open
Abstract
The mechanisms that underlie the regulation of enzymatic reactions by biomolecular condensates and how they scale with compartment size remain poorly understood. Here we use intrinsically disordered domains as building blocks to generate programmable enzymatic condensates of NADH-oxidase (NOX) with different sizes spanning from nanometers to microns. These disordered domains, derived from three distinct RNA-binding proteins, each possessing different net charge, result in the formation of condensates characterized by a comparable high local concentration of the enzyme yet within distinct environments. We show that only condensates with the highest recruitment of substrate and cofactor exhibit an increase in enzymatic activity. Notably, we observe an enhancement in enzymatic rate across a wide range of condensate sizes, from nanometers to microns, indicating that emergent properties of condensates can arise within assemblies as small as nanometers. Furthermore, we show a larger rate enhancement in smaller condensates. Our findings demonstrate the ability of condensates to modulate enzymatic reactions by creating distinct effective solvent environments compared to the surrounding solution, with implications for the design of protein-based heterogeneous biocatalysts.
Collapse
Affiliation(s)
- Marcos Gil-Garcia
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Ana I Benítez-Mateos
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Marcell Papp
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Florence Stoffel
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Chiara Morelli
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Karl Normak
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Katarzyna Makasewicz
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Lenka Faltova
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Francesca Paradisi
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Depenveiller C, Baud S, Belloy N, Bochicchio B, Dandurand J, Dauchez M, Pepe A, Pomès R, Samouillan V, Debelle L. Structural and physical basis for the elasticity of elastin. Q Rev Biophys 2024; 57:e3. [PMID: 38501287 DOI: 10.1017/s0033583524000040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Elastin function is to endow vertebrate tissues with elasticity so that they can adapt to local mechanical constraints. The hydrophobicity and insolubility of the mature elastin polymer have hampered studies of its molecular organisation and structure-elasticity relationships. Nevertheless, a growing number of studies from a broad range of disciplines have provided invaluable insights, and several structural models of elastin have been proposed. However, many questions remain regarding how the primary sequence of elastin (and the soluble precursor tropoelastin) governs the molecular structure, its organisation into a polymeric network, and the mechanical properties of the resulting material. The elasticity of elastin is known to be largely entropic in origin, a property that is understood to arise from both its disordered molecular structure and its hydrophobic character. Despite a high degree of hydrophobicity, elastin does not form compact, water-excluding domains and remains highly disordered. However, elastin contains both stable and labile secondary structure elements. Current models of elastin structure and function are drawn from data collected on tropoelastin and on elastin-like peptides (ELPs) but at the tissue level, elasticity is only achieved after polymerisation of the mature elastin. In tissues, the reticulation of tropoelastin chains in water defines the polymer elastin that bears elasticity. Similarly, ELPs require polymerisation to become elastic. There is considerable interest in elastin especially in the biomaterials and cosmetic fields where ELPs are widely used. This review aims to provide an up-to-date survey of/perspective on current knowledge about the interplay between elastin structure, solvation, and entropic elasticity.
Collapse
Affiliation(s)
- Camille Depenveiller
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens, France
| | - Stéphanie Baud
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Nicolas Belloy
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Brigida Bochicchio
- Laboratory of Bioinspired Materials, Department of Science, University of Basilicata, Potenza, Italy
| | - Jany Dandurand
- CIRIMAT UMR 5085, Université Paul Sabatier, Université de Toulouse, Toulouse, France
| | - Manuel Dauchez
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Antonietta Pepe
- Laboratory of Bioinspired Materials, Department of Science, University of Basilicata, Potenza, Italy
| | - Régis Pomès
- Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Valérie Samouillan
- CIRIMAT UMR 5085, Université Paul Sabatier, Université de Toulouse, Toulouse, France
| | - Laurent Debelle
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
13
|
Eweje F, Walsh ML, Ahmad K, Ibrahim V, Alrefai A, Chen J, Chaikof EL. Protein-based nanoparticles for therapeutic nucleic acid delivery. Biomaterials 2024; 305:122464. [PMID: 38181574 PMCID: PMC10872380 DOI: 10.1016/j.biomaterials.2023.122464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/25/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024]
Abstract
To realize the full potential of emerging nucleic acid therapies, there is a need for effective delivery agents to transport cargo to cells of interest. Protein materials exhibit several unique properties, including biodegradability, biocompatibility, ease of functionalization via recombinant and chemical modifications, among other features, which establish a promising basis for therapeutic nucleic acid delivery systems. In this review, we highlight progress made in the use of non-viral protein-based nanoparticles for nucleic acid delivery in vitro and in vivo, while elaborating on key physicochemical properties that have enabled the use of these materials for nanoparticle formulation and drug delivery. To conclude, we comment on the prospects and unresolved challenges associated with the translation of protein-based nucleic acid delivery systems for therapeutic applications.
Collapse
Affiliation(s)
- Feyisayo Eweje
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard and MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Harvard/MIT MD-PhD Program, Boston, MA, USA, 02115; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Michelle L Walsh
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard and MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Harvard/MIT MD-PhD Program, Boston, MA, USA, 02115
| | - Kiran Ahmad
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Vanessa Ibrahim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Assma Alrefai
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Strader RL, Shmidov Y, Chilkoti A. Encoding Structure in Intrinsically Disordered Protein Biomaterials. Acc Chem Res 2024; 57:302-311. [PMID: 38194282 PMCID: PMC11354101 DOI: 10.1021/acs.accounts.3c00624] [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: 01/10/2024]
Abstract
In nature, proteins range from those with highly ordered secondary and tertiary structures to those that completely lack a well-defined three-dimensional structure, termed intrinsically disordered proteins (IDPs). IDPs are generally characterized by one or more segments that have a compositional bias toward small hydrophilic amino acids and proline residues that promote structural disorder and are called intrinsically disordered regions (IDRs). The combination of IDRs with ordered regions and the interactions between the two determine the phase behavior, structure, and function of IDPs. Nature also diversifies the structure of proteins and thereby their functions by hybridization of the proteins with other moieties such as glycans and lipids; for instance, post-translationally glycosylated and lipidated proteins are important cell membrane components. Additionally, diversity in protein structure and function is achieved in nature through cross-linking proteins within themselves or with other domains to create various topologies. For example, an essential characteristic of the extracellular matrix (ECM) is the cross-linking of its network components, including proteins such as collagen and elastin, as well as polysaccharides such as hyaluronic acid (HA). Inspired by nature, synthetic IDP (SynIDP)-based biomaterials can be designed by employing similar strategies with the goal of introducing structural diversity and hence unique physiochemical properties. This Account describes such materials produced over the past decade and following one or more of the following approaches: (1) incorporating highly ordered domains into SynIDPs, (2) conjugating SynIDPs to other moieties through either genetically encoded post-translational modification or chemical conjugation, and (3) engineering the topology of SynIDPs via chemical modification. These approaches introduce modifications to the primary structure of SynIDPs, which are then translated to unique three-dimensional secondary and tertiary structures. Beginning with completely disordered SynIDPs as the point of origin, structure may be introduced into SynIDPs by each of these three unique approaches individually along orthogonal axes or by combinations of the three, enabling bioinspired designs to theoretically span the entire range of three-dimensional structural possibilities. Furthermore, the resultant structures span a wide range of length scales, from nano- to meso- to micro- and even macrostructures. In this Account, emphasis is placed on the physiochemical properties and structural features of the described materials. Conjugates of SynIDPs to synthetic polymers and materials achieved by simple mixing of components are outside the scope of this Account. Related biomedical applications are described briefly. Finally, we note future directions for the design of functional SynIDP-based biomaterials.
Collapse
Affiliation(s)
- Rachel L. Strader
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708, USA
| | - Yulia Shmidov
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708, USA
| |
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: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
Huang S, Wang K, Hua Z, Abd El-Aty AM, Tan M. Size-controllable food-grade nanoparticles based on sea cucumber polypeptide with good anti-oxidative capacity to prolong lifespan in tumor-bearing mice. Int J Biol Macromol 2023; 253:127039. [PMID: 37742886 DOI: 10.1016/j.ijbiomac.2023.127039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Liver cancer, a malignancy with a rising global incidence, poses a significant challenge in achieving effective treatment outcomes. As food-derived nutrient, sea cucumber peptide (SCP) has shown promising anticancer effects. Therefore, we explored the nanodelivery systems to encapsulate SCP to enhance its stability in the gastrointestinal tract and improve absorption within the tumor microenvironment. This study aimed to develop size-controllable multifunctional nanoparticles using SCP, procyanidins (PCs), and vanillin through molecular assembly via a one-pot Mannich condensation approach. These food-grade nanoparticles demonstrated water solubility and exhibited a spherical structure with sizes ranging from 441 to 1360 nm, depending on the concentration of the reactants. In vitro cell experiments demonstrated that SCP nanoparticles modified with PCs effectively reduced the generation of reactive oxygen species from H2O2 and acrylamide while maintaining normal levels of mitochondrial membrane potential. Furthermore, in vivo nutrition intervention studies conducted on tumor-bearing mice revealed that mice treated with SCP nanoparticles exhibited a survival rate of 40 %, which was significantly higher than the 0 % and 20 % survival rates observed in the control and SCP-treated groups, respectively. These findings suggest that SCP nanoparticles, possessing antioxidative properties and controllable sizes, hold potential for precision nutrition in the field of cancer treatment.
Collapse
Affiliation(s)
- Shasha Huang
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Kuiyou Wang
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Zheng Hua
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25240, Turkey
| | - Mingqian Tan
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
17
|
Suhar RA, Huang MS, Navarro RS, Aviles Rodriguez G, Heilshorn SC. A Library of Elastin-like Proteins with Tunable Matrix Ligands for In Vitro 3D Neural Cell Culture. Biomacromolecules 2023; 24:5926-5939. [PMID: 37988588 DOI: 10.1021/acs.biomac.3c00941] [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: 11/23/2023]
Abstract
Hydrogels with encapsulated cells have widespread biomedical applications, both as tissue-mimetic 3D cultures in vitro and as tissue-engineered therapies in vivo. Within these hydrogels, the presentation of cell-instructive extracellular matrix (ECM)-derived ligands and matrix stiffness are critical factors known to influence numerous cell behaviors. While individual ECM biopolymers can be blended together to alter the presentation of cell-instructive ligands, this typically results in hydrogels with a range of mechanical properties. Synthetic systems that allow for the facile incorporation and modulation of multiple ligands without modification of matrix mechanics are highly desirable. In the present work, we leverage protein engineering to design a family of xeno-free hydrogels (i.e., devoid of animal-derived components) consisting of recombinant hyaluronan and recombinant elastin-like proteins (ELPs), cross-linked together with dynamic covalent bonds. The ELP components incorporate cell-instructive peptide ligands derived from ECM proteins, including fibronectin (RGD), laminin (IKVAV and YIGSR), collagen (DGEA), and tenascin-C (PLAEIDGIELTY and VFDNFVL). By carefully designing the protein primary sequence, we form 3D hydrogels with defined and tunable concentrations of cell-instructive ligands that have similar matrix mechanics. Utilizing this system, we demonstrate that neurite outgrowth from encapsulated embryonic dorsal root ganglion (DRG) cultures is significantly modified by cell-instructive ligand content. Thus, this library of protein-engineered hydrogels is a cell-compatible system to systematically study cell responses to matrix-derived ligands.
Collapse
Affiliation(s)
- Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- The Institute for Chemistry, Stanford University, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford, California 94305, United States
| | - Renato S Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Giselle Aviles Rodriguez
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
18
|
Guo Y, Liu S, Jing D, Liu N, Luo X. The construction of elastin-like polypeptides and their applications in drug delivery system and tissue repair. J Nanobiotechnology 2023; 21:418. [PMID: 37951928 PMCID: PMC10638729 DOI: 10.1186/s12951-023-02184-8] [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: 03/29/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023] Open
Abstract
Elastin-like polypeptides (ELPs) are thermally responsive biopolymers derived from natural elastin. These peptides have a low critical solution temperature phase behavior and can be used to prepare stimuli-responsive biomaterials. Through genetic engineering, biomaterials prepared from ELPs can have unique and customizable properties. By adjusting the amino acid sequence and length of ELPs, nanostructures, such as micelles and nanofibers, can be formed. Correspondingly, ELPs have been used for improving the stability and prolonging drug-release time. Furthermore, ELPs have widespread use in tissue repair due to their biocompatibility and biodegradability. Here, this review summarizes the basic property composition of ELPs and the methods for modulating their phase transition properties, discusses the application of drug delivery system and tissue repair and clarifies the current challenges and future directions of ELPs in applications.
Collapse
Affiliation(s)
- Yingshu Guo
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Shiwei Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Dan Jing
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Nianzu Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Xiliang Luo
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| |
Collapse
|
19
|
Tu J, Liu Q, You S, Meng Z, Fang S, Yu B, Chen X, Zhou Y, Zeng L, Herrmann A, Chen G, Shen J, Zheng L, Ji J. Recombinant supercharged polypeptides for safe and efficient heparin neutralization. Biomater Sci 2023; 11:5533-5539. [PMID: 37395046 DOI: 10.1039/d3bm00628j] [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: 07/04/2023]
Abstract
Heparin is a widely used anticoagulant agent in the clinic. After application, its anticoagulant effect must be reversed to prevent potential side effects. Protamine sulfate (PS) is the only clinically licensed antidote that has been used for this purpose in the last 80 years, which, however, provokes severe adverse effects, such as systemic hypotension and even death. Herein, we demonstrate the potential of supercharged polypeptides as a promising alternative for protamine sulfate. A series of supercharged polypeptides with multiple positive charges was recombinantly produced, and the heparin-neutralizing performance of the polypeptides was evaluated in comparison with PS. It was found that increasing the number of charges significantly enhanced the ability to neutralize heparin and resist the screening effect induced by salt. In particular, the polypeptide bearing 72 charges (K72) exhibited an excellent heparin-neutralizing behavior that was comparable to that of PS. Further in vivo studies revealed that the heparin-triggered bleeding was almost completely alleviated by K72 while a negligible toxic effect was observed. Therefore, such recombinant supercharged polypeptides might replace protamine sulfate as heparin-reversal agents.
Collapse
Affiliation(s)
- Jianfei Tu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China.
| | - Qing Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China.
| | - Shengye You
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China.
| | - Zhuojun Meng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China.
| | - Shiji Fang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China.
| | - Binhong Yu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China.
| | - Xumin Chen
- Department of Nephrology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China
| | - Yu Zhou
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr 50, 52056 Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
| | - Lulu Zeng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China.
| | - Andreas Herrmann
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr 50, 52056 Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074 Aachen, Germany
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianliang Shen
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China.
| | - Lifei Zheng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Imaging Diagnostic and Interventional Minimally Invasive Institute, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang, China.
| |
Collapse
|
20
|
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: 5] [Impact Index Per Article: 2.5] [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
|
21
|
Ma JX, Liu P, Wang YX, Ren X, Zhang R, Li LW. A histidine-rich fusion tag enables real-time monitoring of recombinant protein expression by Pauly reaction-based colorimetric assay. Biochem Biophys Res Commun 2023; 666:128-136. [PMID: 37182288 DOI: 10.1016/j.bbrc.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/16/2023]
Abstract
Commercially available recombinant expression systems always use fusion tags to facilitate target protein purification and SDS-PAGE analysis followed by Coomassie Brilliant Blue (CBB) staining is the classical method to validate the expression level of target protein, which is time-consuming, although not very laborious. Previously, we found that a histidine-rich elastin-like polypeptide (HRELP) tag could make its fusion proteins being quickly and specifically stained with Pauly's reagent. In this study, we designed a Pauly reaction-based colorimetric assay to real-time monitoring of the expression level of recombinant protein tagged HRELP and found that the absorption value of post-induction E. coli cells stained with Pauly's reagent correlated well with both the band intensity of the target protein from Pauly's reagent-stained and CBB-stained gels. Moreover, we found the colorimetric assay could also be helpful to roughly estimate the expression efficiency by using a poly-histidine-tagged protein, which has only 1.17% histidine residue. In our opinion, Pauly reaction-based colorimetric assay could significantly shorten the time to validate the over-expression of recombinant protein tagged with either HRELP or poly-histidine. And HRELP seemed to be an ideal fusion tag for it can not only facilitate protein purification but also simplify protein detection.
Collapse
Affiliation(s)
- Jin-Xuan Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, 710069, China
| | - Pu Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, 710069, China
| | - Yuan-Xiang Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, 710069, China
| | - Xi Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, 710069, China
| | - Rui Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, 710069, China
| | - Li-Wen Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, 710069, China.
| |
Collapse
|
22
|
Tian KK, Qian ZG, Xia XX. Synthetic biology-guided design and biosynthesis of protein polymers for delivery. Adv Drug Deliv Rev 2023; 194:114728. [PMID: 36791475 DOI: 10.1016/j.addr.2023.114728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/28/2022] [Accepted: 02/06/2023] [Indexed: 02/15/2023]
Abstract
Vehicles derived from genetically engineered protein polymers have gained momentum in the field of biomedical engineering due to their unique designability, remarkable biocompatibility and excellent biodegradability. However, the design and production of these protein polymers with on-demand sequences and supramolecular architectures remain underexplored, particularly from a synthetic biology perspective. In this review, we summarize the state-of-the art strategies for constructing the highly repetitive genes encoding the protein polymers, and highlight the advanced approaches for metabolically engineering expression hosts towards high-level biosynthesis of the target protein polymers. Finally, we showcase the typical protein polymers utilized to fabricate delivery vehicles.
Collapse
Affiliation(s)
- Kai-Kai Tian
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Zhi-Gang Qian
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xiao-Xia Xia
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| |
Collapse
|
23
|
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
|
24
|
Garcia Garcia C, Patkar SS, Wang B, Abouomar R, Kiick KL. Recombinant protein-based injectable materials for biomedical applications. Adv Drug Deliv Rev 2023; 193:114673. [PMID: 36574920 DOI: 10.1016/j.addr.2022.114673] [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: 06/08/2022] [Revised: 11/09/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Injectable nanocarriers and hydrogels have found widespread use in a variety of biomedical applications such as local and sustained biotherapeutic cargo delivery, and as cell-instructive matrices for tissue engineering. Recent advances in the development and application of recombinant protein-based materials as injectable platforms under physiological conditions have made them useful platforms for the development of nanoparticles and tissue engineering matrices, which are reviewed in this work. Protein-engineered biomaterials are highly customizable, and they provide distinctly tunable rheological properties, encapsulation efficiencies, and delivery profiles. In particular, the key advantages of emerging technologies which harness the stimuli-responsive properties of recombinant polypeptide-based materials are highlighted in this review.
Collapse
Affiliation(s)
- Cristobal Garcia Garcia
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Sai S Patkar
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Bin Wang
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Ramadan Abouomar
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE 19176, USA.
| |
Collapse
|
25
|
Tailored Functionalized Protein Nanocarriers for Cancer Therapy: Recent Developments and Prospects. Pharmaceutics 2023; 15:pharmaceutics15010168. [PMID: 36678796 PMCID: PMC9861211 DOI: 10.3390/pharmaceutics15010168] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Recently, the potential use of nanoparticles for the targeted delivery of therapeutic and diagnostic agents has garnered increased interest. Several nanoparticle drug delivery systems have been developed for cancer treatment. Typically, protein-based nanocarriers offer several advantages, including biodegradability and biocompatibility. Using genetic engineering or chemical conjugation approaches, well-known naturally occurring protein nanoparticles can be further prepared, engineered, and functionalized in their self-assembly to meet the demands of clinical production efficiency. Accordingly, promising protein nanoparticles have been developed with outstanding tumor-targeting capabilities, ultimately overcoming multidrug resistance issues, in vivo delivery barriers, and mimicking the tumor microenvironment. Bioinspired by natural nanoparticles, advanced computational techniques have been harnessed for the programmable design of highly homogenous protein nanoparticles, which could open new routes for the rational design of vaccines and drug formulations. The current review aims to present several significant advancements made in protein nanoparticle technology, and their use in cancer therapy. Additionally, tailored construction methods and therapeutic applications of engineered protein-based nanoparticles are discussed.
Collapse
|
26
|
Lee K, Noh Y, Bae Y, Kang S, Cha C. Tunable Physicomechanical and Drug Release Properties of In Situ Forming Thermoresponsive Elastin-like Polypeptide Hydrogels. Biomacromolecules 2022; 23:5193-5201. [PMID: 36378752 DOI: 10.1021/acs.biomac.2c01001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
With the continued advancement in the design and engineering of hydrogels for biomedical applications, there is a growing interest in imparting stimuli-responsiveness to the hydrogels in order to control their physicomechanical properties in a more programmable manner. In this study, an in situ forming hydrogel is developed by cross-linking alginate with an elastin-like polypeptide (ELP). Lysine-rich ELP synthesized by recombinant DNA technology is reacted with alginate presenting an aldehyde via Schiff base formation, resulting in facile hydrogel formation under physiological conditions. The physicomechanical properties of alginate-ELP hydrogels can be controlled in a wide range by the concentrations of alginate and ELP. Owing to the thermoresponsive properties of the ELP, the alginate-ELP hydrogels undergo swelling/deswelling near the physiological temperature. Taking advantage of these highly attractive properties of alginate-ELP, drug release kinetics were measured to evaluate their potential as a thermoresponsive drug delivery system. Furthermore, an ex vivo model was used to demonstrate the minimally invasive tissue injectability.
Collapse
Affiliation(s)
- Kangseok Lee
- Center for Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan44919, Republic of Korea
| | - Yeongjin Noh
- Center for Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan44919, Republic of Korea
| | - Yoonji Bae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan44919, Republic of Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan44919, Republic of Korea
| | - Chaenyung Cha
- Center for Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan44919, Republic of Korea
| |
Collapse
|
27
|
The application of elastin-like peptides in cancer, tissue engineering and ocular disease. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
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: 15] [Impact Index Per Article: 5.0] [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
|
29
|
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
|
30
|
Attia SA, MacKay JA. Protein and polypeptide mediated delivery to the eye. Adv Drug Deliv Rev 2022; 188:114441. [PMID: 35817213 PMCID: PMC10049092 DOI: 10.1016/j.addr.2022.114441] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022]
Abstract
Hybrid or recombinant protein-polymers, peptide-based biomaterials, and antibody-targeted therapeutics are widely explored for various ocular conditions and vision correction. They have been noted for their potential biocompatibility, potency, adaptability, and opportunities for sustained drug delivery. Unique to peptide and protein therapeutics, their production by cellular translation allows their precise modification through genetic engineering. To a greater extent than drug delivery to other systems, delivery to the eye can benefit from the combination of locally-targeted administration and protein-based specificity. Consequently, a range of delivery platforms and administration methods have been exploited to address the ocular delivery of peptide and protein biomaterials. This review discusses a sample of preclinical and clinical opportunities for peptide-based drug delivery to the eye.
Collapse
Affiliation(s)
- Sara Aly Attia
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, 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
|
31
|
Avila H, Yu J, Boddu G, Phan A, Truong A, Peddi S, Guo H, Lee SJ, Alba M, Canfield E, Yamamoto V, Paton JC, Paton AW, Lee AS, MacKay JA. Hydra-Elastin-like Polypeptides Increase Rapamycin Potency When Targeting Cell Surface GRP78. Biomacromolecules 2022; 23:3116-3129. [PMID: 35786858 PMCID: PMC10231879 DOI: 10.1021/acs.biomac.2c00048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rapalogues are powerful therapeutic modalities for breast cancer; however, they suffer from low solubility and dose-limiting side effects. To overcome these challenges, we developed a long-circulating multiheaded drug carrier called 5FA, which contains rapamycin-binding domains linked with elastin-like polypeptides (ELPs). To target these "Hydra-ELPs" toward breast cancer, we here linked 5FA with four distinct peptides which are reported to engage the cell surface form of the 78 kDa glucose-regulated protein (csGRP78). To determine if these peptides affected the carrier solubility, this library was characterized by light scattering and mass spectrometry. To guide in vitro selection of the most potent functional carrier for rapamycin, its uptake and inhibition of mTORC1 were monitored in a ductal breast cancer model (BT474). Using flow cytometry to track cellular association, it was found that only the targeted carriers enhanced cellular uptake and were susceptible to proteolysis by SubA, which specifically targets csGRP78. The functional inhibition of mTOR was monitored by Western blot for pS6K, whereby the best carrier L-5FA reduced mTOR activity by 3-fold compared to 5FA or free rapamycin. L-5FA was further visualized using super-resolution confocal laser scanning microscopy, which revealed that targeting increased exposure to the carrier by ∼8-fold. This study demonstrates how peptide ligands for GRP78, such as the L peptide (RLLDTNRPLLPY), may be incorporated into protein-based drug carriers to enhance targeting.
Collapse
Affiliation(s)
- Hugo Avila
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Jingmei Yu
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Geetha Boddu
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Anh Truong
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Santosh Peddi
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Hao Guo
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Shin-Jae Lee
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
- Department of Biomedical Engineering, USC Viterbi School of Engineering, Los Angeles, California 90089, United States
| | - Mario Alba
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Ethan Canfield
- Mass Spectrometry Core, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Vicky Yamamoto
- Department of Biochemistry and Molecular Medicine, USC Keck School of Medicine, Los Angeles, California 90033, United States
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide 5005, Australia
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide 5005, Australia
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, USC Keck School of Medicine, Los Angeles, California 90033, United States
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
- Department of Biomedical Engineering, USC Viterbi School of Engineering, Los Angeles, California 90089, United States
- Department of Ophthalmology, USC Keck School of Medicine, Los Angeles, California 90033, United States
| |
Collapse
|
32
|
Abstract
Protein nanomaterials are well-defined, hollow protein nanoparticles comprised of virus capsids, virus-like particles, ferritin, heat shock proteins, chaperonins and many more. Protein-based nanomaterials are formed by the self-assembly of protein subunits and have numerous desired properties as drug-delivery vehicles, including being optimally sized for endocytosis, nontoxic, biocompatible, biodegradable and functionalized at three separate interfaces (external, internal and intersubunit). As a result, protein nanomaterials have been intensively investigated as functional entities in bionanotechnology, including drug delivery, nanoreactors and templates for organic and inorganic nanomaterials. Several variables influence efficient administration, particularly active targeting, cellular uptake, the kinetics of the release and systemic elimination. This review examines the wide range of medicines, loading/release processes, targeted therapies and treatment effectiveness.
Collapse
|
33
|
Polymer-Based Delivery of Peptide Drugs to Treat Diabetes: Normalizing Hyperglycemia and Preventing Diabetic Complications. BIOCHIP JOURNAL 2022. [DOI: 10.1007/s13206-022-00057-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Kelly G, Milligan JJ, Mastria EM, Kim S, Zelenetz SR, Dobbins J, Cai LY, Li X, Nair SK, Chilkoti A. Intratumoral delivery of brachytherapy and immunotherapy by a thermally triggered polypeptide depot. J Control Release 2022; 343:267-276. [PMID: 35077742 PMCID: PMC8960370 DOI: 10.1016/j.jconrel.2022.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 10/19/2022]
Abstract
Biomaterial-based approaches for a combination of radiotherapy and immunotherapy can improve outcomes in metastatic cancer through local delivery of both therapeutic modalities to the primary tumor to control local tumor growth and distant metastases. This study describes an injectable depot for sustained intratumoral (i.t.) delivery of an iodine-131 (131I) radionuclide and a CpG oligodeoxynucleotide immunostimulant, driven by the thermally sensitive phase transition behavior of elastin-like polypeptides (ELPs). We synthesized and characterized an ELP with an oligolysine tail (ELP-K12) that forms an electrostatic complex with CpG for delivery from an ELP depot and evaluated the ability of the complex to enhance local and systemic tumor control as a monotherapy and in combination with 131I-ELP brachytherapy. I.t delivery of CpG from an ELP-K12 depot dramatically prolongs i.t. retention to more than 21 days as compared to soluble CpG that is only retained within the tumor for <24 h. ELP-K12 also enhances CpG delivery by increasing cellular uptake of CpG to generate greater toll-like receptor 9 (TLR9) activation than CpG alone. I.t. treatment with an ELP-K12/CpG depot slows primary tumor growth and reduces lung metastases in a poorly immunogenic 4 T1 syngeneic breast cancer model whereas i.t treatment of CpG alone has no significant effect on primary tumor growth or metastases. Notably, a combination of 131I-ELP brachytherapy and ELP-K12/CpG delivered i.t. inhibited 4 T1 tumor growth and strongly decreased the development of lung metastases, leading to a synergistic improvement in mouse survival. These preclinical results demonstrate that injectable ELP depots may provide a useful approach for the delivery of combination radio- and immuno-therapy to treat metastatic disease.
Collapse
Affiliation(s)
- Garrett Kelly
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Joshua J. Milligan
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Eric M. Mastria
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Sarah Kim
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Stephanie R. Zelenetz
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Jarrett Dobbins
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Leon Y. Cai
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Xinghai Li
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Smita K. Nair
- Department of Surgery, Duke University School of Medicine, 2301 Erwin Rd., DUMC Box 370, Durham, NC 27710, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA.
| |
Collapse
|
35
|
Genetically encoded elastin-like polypeptide nanoparticles for drug delivery. Curr Opin Biotechnol 2021; 74:146-153. [PMID: 34920210 DOI: 10.1016/j.copbio.2021.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 12/22/2022]
Abstract
Small molecule drugs suffer from poor in vivo half-life, rapid degradation, and systemic off-target toxicity. To address these issues, researchers have developed nanoparticles that significantly enhance the delivery of many drugs while reducing their toxicity and improving targeting to specific organs. Recombinantly synthesized biomaterials such as elastin-like polypeptides (ELPs) have unique attributes that greatly facilitate the rational design of nanoparticles for drug delivery. These attributes include biocompatibility, precise control over amino acid sequence design, and stimuli-responsive self-assembly into nanostructures that can be loaded with a range of drugs to enhance their pharmacokinetics and pharmacodynamics, significantly improving their therapeutic efficacy over the free drugs. This review summarizes recent developments in genetically encoded, self-assembling ELP nanoparticles and their applications for drug delivery.
Collapse
|
36
|
Dunshee LC, Sullivan MO, Kiick KL. Therapeutic nanocarriers comprising extracellular matrix-inspired peptides and polysaccharides. Expert Opin Drug Deliv 2021; 18:1723-1740. [PMID: 34696691 PMCID: PMC8601199 DOI: 10.1080/17425247.2021.1988925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The extracellular matrix (ECM) is vital for cell and tissue development. Given its importance, extensive work has been conducted to develop biomaterials and drug delivery vehicles that capture features of ECM structure and function. AREAS COVERED This review highlights recent developments of ECM-inspired nanocarriers and their exploration for drug and gene delivery applications. Nanocarriers that are inspired by or created from primary components of the ECM (e.g. elastin, collagen, hyaluronic acid (HA), or combinations of these) are explicitly covered. An update on current clinical trials employing elastin-like proteins is also included. EXPERT OPINION Novel ECM-inspired nanoscale structures and conjugates continue to be of great interest in the materials science and bioengineering communities. Hyaluronic acid nanocarrier systems in particular are widely employed due to the functional activity of HA in mediating a large number of disease states. In contrast, collagen-like peptide nanocarriers are an emerging drug delivery platform with potential relevance to a myriad of ECM-related diseases, making their continued study most pertinent. Elastin-like peptide nanocarriers have a well-established tolerability and efficacy track record in preclinical analyses that has motivated their recent advancement into the clinical arena.
Collapse
Affiliation(s)
- Lucas C Dunshee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Kristi L Kiick
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|