1
|
Bojarska J, Wolf WM. Short Peptides as Powerful Arsenal for Smart Fighting Cancer. Cancers (Basel) 2024; 16:3254. [PMID: 39409876 PMCID: PMC11476321 DOI: 10.3390/cancers16193254] [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: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Short peptides have been coming around as a strong weapon in the fight against cancer on all fronts-in immuno-, chemo-, and radiotherapy, and also in combinatorial approaches. Moreover, short peptides have relevance in cancer imaging or 3D culture. Thanks to the natural 'smart' nature of short peptides, their unique structural features, as well as recent progress in biotechnological and bioinformatics development, short peptides are playing an enormous role in evolving cutting-edge strategies. Self-assembling short peptides may create excellent structures to stimulate cytotoxic immune responses, which is essential for cancer immunotherapy. Short peptides can help establish versatile strategies with high biosafety and effectiveness. Supramolecular short peptide-based cancer vaccines entered clinical trials. Peptide assemblies can be platforms for the delivery of antigens, adjuvants, immune cells, and/or drugs. Short peptides have been unappreciated, especially in the vaccine aspect. Meanwhile, they still hide the undiscovered unlimited potential. Here, we provide a timely update on this highly active and fast-evolving field.
Collapse
Affiliation(s)
- Joanna Bojarska
- Chemistry Department, Institute of Inorganic and Ecological Chemistry, Łódź University of Technology, S. Żeromskiego Str. 116, 90-924 Łódź, Poland;
| | | |
Collapse
|
2
|
Li Z, Song K, Chen Y, Huang Q, You L, Yu L, Chen B, Yuan Z, Xu Y, Su Y, Da L, Zhu X, Dong R. Sequence-encoded bioactive protein-multiblock polymer conjugates via quantitative one-pot iterative living polymerization. Nat Commun 2024; 15:6729. [PMID: 39112493 PMCID: PMC11306232 DOI: 10.1038/s41467-024-51122-1] [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: 09/15/2023] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Protein therapeutics are essential in treating various diseases, but their inherent biological instability and short circulatory half-lives in vivo pose challenges. Herein, a quantitative one-pot iterative living polymerization technique is reported towards precision control over the molecular structure and monomer sequence of protein-polymer conjugates, aiming to maximize physicochemical properties and biological functions of proteins. Using this quantitative one-pot iterative living polymerization technique, we successfully develop a series of sequence-controlled protein-multiblock polymer conjugates, enhancing their biostability, pharmacokinetics, cellular uptake, and in vivo biodistribution. All-atom molecular dynamics simulations are performed to disclose the definite sequence-function relationship of the bioconjugates, further demonstrating their sequence-encoded cellular uptake behavior and in vivo biodistribution in mice. Overall, this work provides a robust approach for creating precision protein-polymer conjugates with defined sequences and advanced functions as a promising candidate in disease treatment.
Collapse
Affiliation(s)
- Ziying Li
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Kaiyuan Song
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yu Chen
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Qijing Huang
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Lujia You
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Li Yu
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Baiyang Chen
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Zihang Yuan
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- School of Chemistry and Chemical Engineering, Frontiers Science Centre for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, China
| | - Yaqin Xu
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yue Su
- School of Chemistry and Chemical Engineering, Frontiers Science Centre for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, China
| | - Lintai Da
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Centre for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, China
| | - Ruijiao Dong
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
3
|
Khakpour S, Hosano N, Moosavi-Nejad Z, Farajian AA, Hosano H. Advancing Tumor Therapy: Development and Utilization of Protein-Based Nanoparticles. Pharmaceutics 2024; 16:887. [PMID: 39065584 PMCID: PMC11279530 DOI: 10.3390/pharmaceutics16070887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Protein-based nanoparticles (PNPs) in tumor therapy hold immense potential, combining targeted delivery, minimal toxicity, and customizable properties, thus paving the way for innovative approaches to cancer treatment. Understanding the various methods available for their production is crucial for researchers and scientists aiming to harness these nanoparticles for diverse applications, including tumor therapy, drug delivery, imaging, and tissue engineering. This review delves into the existing techniques for producing PNPs and PNP/drug complexes, while also exploring alternative novel approaches. The methods outlined in this study were divided into three key categories based on their shared procedural steps: solubility change, solvent substitution, and thin flow methods. This classification simplifies the understanding of the underlying mechanisms by offering a clear framework, providing several advantages over other categorizations. The review discusses the principles underlying each method, highlighting the factors influencing the nanoparticle size, morphology, stability, and functionality. It also addresses the challenges and considerations associated with each method, including the scalability, reproducibility, and biocompatibility. Future perspectives and emerging trends in PNPs' production are discussed, emphasizing the potential for innovative strategies to overcome current limitations, which will propel the field forward for biomedical and therapeutic applications.
Collapse
Affiliation(s)
- Shirin Khakpour
- Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan;
| | - Nushin Hosano
- Department of Biomaterials and Bioelectrics, Institute of Industrial Nanomaterials, Kumamoto University, Kumamoto 860-8555, Japan;
| | - Zahra Moosavi-Nejad
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran 1993893973, Iran
| | - Amir A. Farajian
- Department of Mechanical and Materials Engineering, Wright State University, Dayton, OH 45435, USA;
| | - Hamid Hosano
- Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan;
- Department of Biomaterials and Bioelectrics, Institute of Industrial Nanomaterials, Kumamoto University, Kumamoto 860-8555, Japan;
| |
Collapse
|
4
|
Adams SC, Nambiar AK, Bressler EM, Raut CP, Colson YL, Wong WW, Grinstaff MW. Immunotherapies for locally aggressive cancers. Adv Drug Deliv Rev 2024; 210:115331. [PMID: 38729264 PMCID: PMC11228555 DOI: 10.1016/j.addr.2024.115331] [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: 02/06/2024] [Revised: 03/31/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Improving surgical resection outcomes for locally aggressive tumors is key to inducing durable locoregional disease control and preventing progression to metastatic disease. Macroscopically complete resection of the tumor is the standard of care for many cancers, including breast, ovarian, lung, sarcoma, and mesothelioma. Advancements in cancer diagnostics are increasing the number of surgically eligible cases through early detection. Thus, a unique opportunity arises to improve patient outcomes with decreased recurrence rates via intraoperative delivery treatments using local drug delivery strategies after the tumor has been resected. Of the current systemic treatments (e.g., chemotherapy, targeted therapies, and immunotherapies), immunotherapies are the latest approach to offer significant benefits. Intraoperative strategies benefit from direct access to the tumor microenvironment which improves drug uptake to the tumor and simultaneously minimizes the risk of drug entering healthy tissues thereby resulting in fewer or less toxic adverse events. We review the current state of immunotherapy development and discuss the opportunities that intraoperative treatment provides. We conclude by summarizing progress in current research, identifying areas for exploration, and discussing future prospects in sustained remission.
Collapse
Affiliation(s)
- Sarah C Adams
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Arun K Nambiar
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Eric M Bressler
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yolonda L Colson
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston MA 02215, USA.
| |
Collapse
|
5
|
Battaglini M, Marino A, Montorsi M, Carmignani A, Ceccarelli MC, Ciofani G. Nanomaterials as Microglia Modulators in the Treatment of Central Nervous System Disorders. Adv Healthc Mater 2024; 13:e2304180. [PMID: 38112345 DOI: 10.1002/adhm.202304180] [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: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Microglia play a pivotal role in the central nervous system (CNS) homeostasis, acting as housekeepers and defenders of the surrounding environment. These cells can elicit their functions by shifting into two main phenotypes: pro-inflammatory classical phenotype, M1, and anti-inflammatory alternative phenotype, M2. Despite their pivotal role in CNS homeostasis, microglia phenotypes can influence the development and progression of several CNS disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, ischemic stroke, traumatic brain injuries, and even brain cancer. It is thus clear that the possibility of modulating microglia activation has gained attention as a therapeutic tool against many CNS pathologies. Nanomaterials are an unprecedented tool for manipulating microglia responses, in particular, to specifically target microglia and elicit an in situ immunomodulation activity. This review focuses the discussion on two main aspects: analyzing the possibility of using nanomaterials to stimulate a pro-inflammatory response of microglia against brain cancer and introducing nanostructures able to foster an anti-inflammatory response for treating neurodegenerative disorders. The final aim is to stimulate the analysis of the development of new microglia nano-immunomodulators, paving the way for innovative and effective therapeutic approaches for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Margherita Montorsi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Alessio Carmignani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Maria Cristina Ceccarelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| |
Collapse
|
6
|
Herpoldt K, López CL, Sappington I, Pham MN, Srinivasan S, Netland J, Montgomery KS, Roy D, Prossnitz AN, Ellis D, Wargacki AJ, Pepper M, Convertine AJ, Stayton PS, King NP. Macromolecular Cargo Encapsulation via In Vitro Assembly of Two-Component Protein Nanoparticles. Adv Healthc Mater 2024; 13:e2303910. [PMID: 38180445 PMCID: PMC11468305 DOI: 10.1002/adhm.202303910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/19/2023] [Indexed: 01/06/2024]
Abstract
Self-assembling protein nanoparticles are a promising class of materials for targeted drug delivery. Here, the use of a computationally designed, two-component, icosahedral protein nanoparticle is reported to encapsulate multiple macromolecular cargoes via simple and controlled self-assembly in vitro. Single-stranded RNA molecules between 200 and 2500 nucleotides in length are encapsulated and protected from enzymatic degradation for up to a month with length-dependent decay rates. Immunogenicity studies of nanoparticles packaging synthetic polymers carrying a small-molecule TLR7/8 agonist show that co-delivery of antigen and adjuvant results in a more than 20-fold increase in humoral immune responses while minimizing systemic cytokine secretion associated with free adjuvant. Coupled with the precise control over nanoparticle structure offered by computational design, robust and versatile encapsulation via in vitro assembly opens the door to a new generation of cargo-loaded protein nanoparticles that can combine the therapeutic effects of multiple drug classes.
Collapse
Affiliation(s)
- Karla‐Luise Herpoldt
- Department of BiochemistryUniversity of WashingtonSeattleWA98195USA
- Institute for Protein DesignUniversity of WashingtonSeattleWA98195USA
- Present address:
2seventy BioSeattleWA98102USA
| | - Ciana L. López
- Department of BioengineeringUniversity of WashingtonSeattleWA98195USA
| | - Isaac Sappington
- Department of BiochemistryUniversity of WashingtonSeattleWA98195USA
- Institute for Protein DesignUniversity of WashingtonSeattleWA98195USA
| | - Minh N. Pham
- Department of BiochemistryUniversity of WashingtonSeattleWA98195USA
- Institute for Protein DesignUniversity of WashingtonSeattleWA98195USA
| | - Selvi Srinivasan
- Department of BioengineeringUniversity of WashingtonSeattleWA98195USA
| | - Jason Netland
- Department of ImmunologyUniversity of WashingtonSeattleWA98195USA
| | | | - Debashish Roy
- Department of BioengineeringUniversity of WashingtonSeattleWA98195USA
| | | | - Daniel Ellis
- Department of BiochemistryUniversity of WashingtonSeattleWA98195USA
- Institute for Protein DesignUniversity of WashingtonSeattleWA98195USA
| | - Adam J. Wargacki
- Department of BiochemistryUniversity of WashingtonSeattleWA98195USA
- Institute for Protein DesignUniversity of WashingtonSeattleWA98195USA
| | - Marion Pepper
- Department of ImmunologyUniversity of WashingtonSeattleWA98195USA
| | - Anthony J. Convertine
- Department of BioengineeringUniversity of WashingtonSeattleWA98195USA
- Present address:
Department of Material Science and EngineeringMissouri University of Science and TechnologyRollaMO65409USA
| | | | - Neil P. King
- Department of BiochemistryUniversity of WashingtonSeattleWA98195USA
- Institute for Protein DesignUniversity of WashingtonSeattleWA98195USA
| |
Collapse
|
7
|
Huang X, Zhu X, Yang H, Li Q, Gai L, Sui X, Lu H, Feng J. Nanomaterial Delivery Vehicles for the Development of Neoantigen Tumor Vaccines for Personalized Treatment. Molecules 2024; 29:1462. [PMID: 38611742 PMCID: PMC11012694 DOI: 10.3390/molecules29071462] [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: 02/22/2024] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Tumor vaccines have been considered a promising therapeutic approach for treating cancer in recent years. With the development of sequencing technologies, tumor vaccines based on neoantigens or genomes specifically expressed in tumor cells, mainly in the form of peptides, nucleic acids, and dendritic cells, are beginning to receive widespread attention. Therefore, in this review, we have introduced different forms of neoantigen vaccines and discussed the development of these vaccines in treating cancer. Furthermore, neoantigen vaccines are influenced by factors such as antigen stability, weak immunogenicity, and biosafety in addition to sequencing technology. Hence, the biological nanomaterials, polymeric nanomaterials, inorganic nanomaterials, etc., used as vaccine carriers are principally summarized here, which may contribute to the design of neoantigen vaccines for improved stability and better efficacy.
Collapse
Affiliation(s)
- Xiaoyu Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Xiaolong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Huan Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Qinyi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Lizhi Gai
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| | - Hua Lu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, and Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China;
| | - Jiao Feng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (X.H.); (X.Z.); (H.Y.); (Q.L.); (X.S.)
| |
Collapse
|
8
|
Xu H, Mao B, Ni S, Xie X, Tang S, Wang Y, Zan X, Zheng Q, Huang W. Engineering Matrix-Free Drug Protein Nanoparticles with Promising Penetration through Biobarriers for Treating Corneal Neovascularization. ACS NANO 2024; 18:8209-8228. [PMID: 38452114 DOI: 10.1021/acsnano.3c12203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Protein drugs have been widely used in treating various clinical diseases because of their high specificity, fewer side effects, and favorable therapeutic effect, but they greatly suffer from their weak permeability through tissue barriers, high sensitivity to microenvironments, degradation by proteases, and rapid clearance by the immune system. Herein, we disrupted the standard protocol where protein drugs must be delivered as the cargo via a delivery system and innovatively developed a free entrapping matrix strategy by simply mixing bevacizumab (Beva) with zinc ions to generate Beva-NPs (Beva-Zn2+), where Beva is coordinatively cross-linked by zinc ions with a loading efficiency as high as 99.2% ± 0.41%. This strategy was universal to generating various protein NPs, with different metal ions (Cu2+, Fe3+, Mg2+, Sr2+). The synthetic conditions of Beva-NPs were optimized, and the generated mechanism was investigated in detail. The entrapment, releasing profile, and the bioactivities of released Beva were thoroughly studied. By using in situ doping of the fourth-generation polyamindoamine dendrimer (G4), the Beva-G4-NPs exhibited extended ocular retention and penetration through biobarriers in the anterior segment through transcellular and paracellular pathways, effectively inhibiting corneal neovascularization (CNV) from 91.6 ± 2.03% to 13.5 ± 1.87% in a rat model of CNV. This study contributes to engineering of protein NPs by using a facile strategy for overcoming the weaknesses of protein drugs and protein NPs, such as weak tissue barrier permeability, low encapsulation efficiency, poor loading capacity, and susceptibility to inactivation.
Collapse
Affiliation(s)
- Hongyan Xu
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, People's Republic of China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, People's Republic of China
| | - Bangxun Mao
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, People's Republic of China
| | - Shulan Ni
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, People's Republic of China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, People's Republic of China
| | - Xiaoling Xie
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, People's Republic of China
| | - Sicheng Tang
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, People's Republic of China
| | - Yang Wang
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, People's Republic of China
| | - Xingjie Zan
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, People's Republic of China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, People's Republic of China
| | - Qinxiang Zheng
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, People's Republic of China
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo 315000, People's Republic of China
| | - Wenjuan Huang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, People's Republic of China
| |
Collapse
|
9
|
Li Y, Zhang W, Tang C, Wang C, Liu C, Chen Q, Yang K, Gu Y, Lei P, Xu H, Wang R. Antidiabetic effects and mechanism of γ-polyglutamic acid on type II diabetes mice. Int J Biol Macromol 2024; 261:129809. [PMID: 38290633 DOI: 10.1016/j.ijbiomac.2024.129809] [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: 12/11/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Diabetes is one of the foremost chronic non-communicable diseases worldwide, which significantly impacts people's quality of life. This study aimed to investigate the hypoglycemic effects of γ-polyglutamic acid (γ-PGA) on STZ-induced type II diabetes mice and its potential mechanisms. The results indicated that γ-PGA intervention contributed to reducing fasting blood glucose levels in diabetic mice, regulating lipid metabolism in type II diabetes mice, and improving insulin resistance. Additionally, γ-PGA could alleviate liver inflammation, enhancing the activity of hepatic antioxidant enzymes. Investigation into the insulin signaling pathway revealed that γ-PGA significantly increased the expression of INSR, IRS-1, Akt, PI3K in diabetic mice, thereby enhancing insulin sensitivity and improving insulin resistance to regulate glucose metabolism. High-throughput sequencing of mouse gut microbiota using 16S rRNA showed that γ-PGA increased the abundance and evenness of beneficial bacteria in the intestines of type II diabetic mice, inhibited the growth of harmful bacteria, and may exerted hypoglycemic effects by modulating and improving relevant metabolic pathways associated with diabetes symptoms. This study provides new insights into the treatment of type II diabetes and highlights the significant potential of γ-PGA in treating type II diabetes.
Collapse
Affiliation(s)
- Ying Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Weijie Zhang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Chao Tang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Chen Wang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Changhui Liu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Qian Chen
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Kai Yang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yian Gu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Peng Lei
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Hong Xu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Rui Wang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China; State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
10
|
Kim K, Kim G, Bae J, Song J, Kim H. A pH-Responsive Virus-Like Particle as a Protein Cage for a Targeted Delivery. Adv Healthc Mater 2024; 13:e2302656. [PMID: 37966427 PMCID: PMC11469083 DOI: 10.1002/adhm.202302656] [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: 08/13/2023] [Revised: 11/05/2023] [Indexed: 11/16/2023]
Abstract
A stimuli-responsive protein self-assembly offers promising utility as a protein nanocage for biotechnological and medical applications. Herein, the development of a virus-like particle (VLP) that undergoes a transition between assembly and disassembly under a neutral and acidic pH, respectively, for a targeted delivery is reported. The structure of the bacteriophage P22 coat protein is used for the computational design of coat subunits that self-assemble into a pH-responsive VLP. Subunit designs are generated through iterative computational cycles of histidine substitutions and evaluation of the interaction energies among the subunits under an acidic and neutral pH. The top subunit designs are tested and one that is assembled into a VLP showing the highest pH-dependent structural transition is selected. The cryo-EM structure of the VLP is determined, and the structural basis of a pH-triggered disassembly is delineated. The utility of the designed VLP is exemplified through the targeted delivery of a cytotoxic protein cargo into tumor cells in a pH-dependent manner. These results provide strategies for the development of self-assembling protein architectures with new functionality for diverse applications.
Collapse
Affiliation(s)
- Kwan‐Jip Kim
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Gijeong Kim
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Jin‐Ho Bae
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Ji‐Joon Song
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Hak‐Sung Kim
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| |
Collapse
|
11
|
Shin J, Cole BD, Seyedmohammad M, Lim SI, Jang Y. Protein Nanocarriers Capable of Encapsulating Both Hydrophobic and Hydrophilic Drugs. Methods Mol Biol 2024; 2720:143-150. [PMID: 37775663 DOI: 10.1007/978-1-0716-3469-1_10] [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: 10/01/2023]
Abstract
Protein nanoparticles are promising targeted drug delivery carriers due to their low toxicity, biodegradability, and abundance of proteins in natural sources. Also, protein nanoparticles enable surface modification with other functional proteins or carbohydrate ligands, which improves the efficacy of targeted drug delivery. Nonetheless, a persistent challenge remains to make versatile protein nanoparticles that deliver diverse types of drugs in a wide range of water solubility. Herein, we describe the methods to fabricate nanoparticles made from bovine serum albumin (BSA) that allow for the encapsulation of both hydrophilic and hydrophobic drug molecules, doxorubicin and bilirubin, respectively.
Collapse
Affiliation(s)
- Jooyong Shin
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Blair D Cole
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | | | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Busan, Republic of Korea
| | - Yeongseon Jang
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Tartari APS, Peczek SH, Fin MT, Ziebarth J, Machado CS, Mainardes RM. Bovine Serum Albumin Nanoparticles Enhanced the Intranasal Bioavailability of Silybin in Rats. Pharmaceutics 2023; 15:2648. [PMID: 38139990 PMCID: PMC10747608 DOI: 10.3390/pharmaceutics15122648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Silybin (SLB), an important flavonoid from silymarin, displays significant hepatoprotective, anticancer, antioxidant, and neuroprotective effects. However, its therapeutic efficacy is limited by its low solubility and bioavailability. To address these challenges, we engineered bovine serum albumin (BSA) nanoparticles (NP) loaded with SLB (BSA-NP/SLB) using the coacervation method. BSA-SLB NP exhibited a spherical shape, a mean size of 197 nm, a polydispersity index of 0.275, a zeta potential of -34 mV, and an entrapment efficiency of 67%. X-ray diffraction analysis indicated amorphization of SLB upon encapsulation. Formulation stability was upheld over 180 days. In vitro release assays demonstrated controlled diffusion-erosion release, with approximately 40% SLB released within 0.5 h and 100% over 12 h. Intranasal administration of BSA-NP/SLB in rats improved SLB bioavailability by fourfold compared to free SLB. These findings highlight the promising potential of intranasally administered BSA-NP/SLB as an alternative approach to enhance SLB bioavailability, paving the way for innovative therapeutic applications.
Collapse
Affiliation(s)
- Ana Paula Santos Tartari
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia St., 838, Guarapuava 85040-167, Brazil
| | - Samila Horst Peczek
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia St., 838, Guarapuava 85040-167, Brazil
| | - Margani Taise Fin
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia St., 838, Guarapuava 85040-167, Brazil
| | - Jeferson Ziebarth
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia St., 838, Guarapuava 85040-167, Brazil
| | - Christiane Schineider Machado
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia St., 838, Guarapuava 85040-167, Brazil
| | - Rubiana Mara Mainardes
- Laboratory of Nanostructured Formulations, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia St., 838, Guarapuava 85040-167, Brazil
- Department of Pharmacy, Universidade Estadual do Centro-Oeste, Alameda Élio Antonio Dalla Vecchia St., 838, Guarapuava 85040-167, Brazil
| |
Collapse
|
13
|
Kaltbeitzel J, Wich PR. Protein-based Nanoparticles: From Drug Delivery to Imaging, Nanocatalysis and Protein Therapy. Angew Chem Int Ed Engl 2023; 62:e202216097. [PMID: 36917017 DOI: 10.1002/anie.202216097] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/16/2023]
Abstract
Proteins and enzymes are versatile biomaterials for a wide range of medical applications due to their high specificity for receptors and substrates, high degradability, low toxicity, and overall good biocompatibility. Protein nanoparticles are formed by the arrangement of several native or modified proteins into nanometer-sized assemblies. In this review, we will focus on artificial nanoparticle systems, where proteins are the main structural element and not just an encapsulated payload. While under natural conditions, only certain proteins form defined aggregates and nanoparticles, chemical modifications or a change in the physical environment can further extend the pool of available building blocks. This allows the assembly of many globular proteins and even enzymes. These advances in preparation methods led to the emergence of new generations of nanosystems that extend beyond transport vehicles to diverse applications, from multifunctional drug delivery to imaging, nanocatalysis and protein therapy.
Collapse
Affiliation(s)
- Jonas Kaltbeitzel
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
14
|
Sharda D, Kaur P, Choudhury D. Protein-modified nanomaterials: emerging trends in skin wound healing. DISCOVER NANO 2023; 18:127. [PMID: 37843732 PMCID: PMC10579214 DOI: 10.1186/s11671-023-03903-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 09/23/2023] [Indexed: 10/17/2023]
Abstract
Prolonged inflammation can impede wound healing, which is regulated by several proteins and cytokines, including IL-4, IL-10, IL-13, and TGF-β. Concentration-dependent effects of these molecules at the target site have been investigated by researchers to develop them as wound-healing agents by regulating signaling strength. Nanotechnology has provided a promising approach to achieve tissue-targeted delivery and increased effective concentration by developing protein-functionalized nanoparticles with growth factors (EGF, IGF, FGF, PDGF, TGF-β, TNF-α, and VEGF), antidiabetic wound-healing agents (insulin), and extracellular proteins (keratin, heparin, and silk fibroin). These molecules play critical roles in promoting cell proliferation, migration, ECM production, angiogenesis, and inflammation regulation. Therefore, protein-functionalized nanoparticles have emerged as a potential strategy for improving wound healing in delayed or impaired healing cases. This review summarizes the preparation and applications of these nanoparticles for normal or diabetic wound healing and highlights their potential to enhance wound healing.
Collapse
Affiliation(s)
- Deepinder Sharda
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Pawandeep Kaur
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Diptiman Choudhury
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Thapar Institute of Engineering and Technology-Virginia Tech Centre of Excellence for Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
15
|
Burlec AF, Corciova A, Boev M, Batir-Marin D, Mircea C, Cioanca O, Danila G, Danila M, Bucur AF, Hancianu M. Current Overview of Metal Nanoparticles' Synthesis, Characterization, and Biomedical Applications, with a Focus on Silver and Gold Nanoparticles. Pharmaceuticals (Basel) 2023; 16:1410. [PMID: 37895881 PMCID: PMC10610223 DOI: 10.3390/ph16101410] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Metal nanoparticles (NPs) have garnered considerable attention, due to their unique physicochemical properties, that render them promising candidates for various applications in medicine and industry. This article offers a comprehensive overview of the most recent advancements in the manufacturing, characterization, and biomedical utilization of metal NPs, with a primary focus on silver and gold NPs. Their potential as effective anticancer, anti-inflammatory, and antimicrobial agents, drug delivery systems, and imaging agents in the diagnosis and treatment of a variety of disorders is reviewed. Moreover, their translation to therapeutic settings, and the issue of their inclusion in clinical trials, are assessed in light of over 30 clinical investigations that concentrate on administering either silver or gold NPs in conditions ranging from nosocomial infections to different types of cancers. This paper aims not only to examine the biocompatibility of nanomaterials but also to emphasize potential challenges that may limit their safe integration into healthcare practices. More than 100 nanomedicines are currently on the market, which justifies ongoing study into the use of nanomaterials in medicine. Overall, the present review aims to highlight the potential of silver and gold NPs as innovative and effective therapeutics in the field of biomedicine, citing some of their most relevant current applications.
Collapse
Affiliation(s)
- Ana Flavia Burlec
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Andreia Corciova
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Monica Boev
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Denisa Batir-Marin
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Cornelia Mircea
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Oana Cioanca
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| | - Gabriela Danila
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Marius Danila
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Anca Florentina Bucur
- Research Centre in the Medical-Pharmaceutical Field, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.D.); (M.D.); (A.F.B.)
| | - Monica Hancianu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (A.F.B.); (A.C.); (C.M.); (O.C.); (M.H.)
| |
Collapse
|
16
|
Malek-Khatabi A, Sadat Razavi M, Abdollahi A, Rahimzadeghan M, Moammeri F, Sheikhi M, Tavakoli M, Rad-Malekshahi M, Faraji Rad Z. Recent progress in PLGA-based microneedle-mediated transdermal drug and vaccine delivery. Biomater Sci 2023; 11:5390-5409. [PMID: 37387317 DOI: 10.1039/d3bm00795b] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Microneedles (MNs) have recently been found to have applications in drug, vitamin, protein and vaccine delivery. Polymeric MN arrays continue to attract increasing attention due to their capability to bypass the skin's stratum corneum (SC) barrier with minimal invasiveness. These carriers can achieve the targeted intradermal delivery of drugs and vaccines and improve their transdermal delivery level. As a nontoxic FDA-approved copolymer, polylactic glycolic acid (PLGA) has good biocompatibility and biodegradability. Currently, PLGA-based MNs have a noticeable tendency to be utilized as a delivery system. This study focuses on the most recent advances in PLGA-based MNs. Both PLGA nanoparticle-based MNs and PLGA matrix-based MNs, created for the delivery of vaccines, drugs, proteins and other therapeutic agents, are discussed. The paper also discusses the various types of MNs and their potential applications. Finally, the prospects and challenges of PLGA-based MNs are reviewed.
Collapse
Affiliation(s)
- Atefeh Malek-Khatabi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Malihe Sadat Razavi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alyeh Abdollahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Rahimzadeghan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moammeri
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Sheikhi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamadreza Tavakoli
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Faraji Rad
- School of Engineering, University of Southern Queensland, Springfield, QLD 4300, Australia.
| |
Collapse
|
17
|
Tan JS, Jaffar Ali MNB, Gan BK, Tan WS. Next-generation viral nanoparticles for targeted delivery of therapeutics: Fundamentals, methods, biomedical applications, and challenges. Expert Opin Drug Deliv 2023; 20:955-978. [PMID: 37339432 DOI: 10.1080/17425247.2023.2228202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION Viral nanoparticles (VNPs) are virus-based nanocarriers that have been studied extensively and intensively for biomedical applications. However, their clinical translation is relatively low compared to the predominating lipid-based nanoparticles. Therefore, this article describes the fundamentals, challenges, and solutions of the VNP-based platform, which will leverage the development of next-generation VNPs. AREAS COVERED Different types of VNPs and their biomedical applications are reviewed comprehensively. Strategies and approaches for cargo loading and targeted delivery of VNPs are examined thoroughly. The latest developments in controlled release of cargoes from VNPs and their mechanisms are highlighted too. The challenges faced by VNPs in biomedical applications are identified, and solutions are provided to overcome them. EXPERT OPINION In the development of next-generation VNPs for gene therapy, bioimaging and therapeutic deliveries, focus must be given to reduce their immunogenicity, and increase their stability in the circulatory system. Modular virus-like particles (VLPs) which are produced separately from their cargoes or ligands before all the components are coupled can speed up clinical trials and commercialization. In addition, removal of contaminants from VNPs, cargo delivery across the blood brain barrier (BBB), and targeting of VNPs to organelles intracellularly are challenges that will preoccupy researchers in this decade.
Collapse
Affiliation(s)
- Jia Sen Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Muhamad Norizwan Bin Jaffar Ali
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Bee Koon Gan
- Department of Biological Science, Faculty of Science, National University of Singapore, Singapore
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
18
|
Proteins and their functionalization for finding therapeutic avenues in cancer: Current status and future prospective. Biochim Biophys Acta Rev Cancer 2023; 1878:188862. [PMID: 36791920 DOI: 10.1016/j.bbcan.2023.188862] [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: 06/24/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Despite the remarkable advancement in the health care sector, cancer remains the second most fatal disease globally. The existing conventional cancer treatments primarily include chemotherapy, which has been associated with little to severe side effects, and radiotherapy, which is usually expensive. To overcome these problems, target-specific nanocarriers have been explored for delivering chemo drugs. However, recent reports on using a few proteins having anticancer activity and further use of them as drug carriers have generated tremendous attention for furthering the research towards cancer therapy. Biomolecules, especially proteins, have emerged as suitable alternatives in cancer treatment due to multiple favourable properties including biocompatibility, biodegradability, and structural flexibility for easy surface functionalization. Several in vitro and in vivo studies have reported that various proteins derived from animal, plant, and bacterial species, demonstrated strong cytotoxic and antiproliferative properties against malignant cells in native and their different structural conformations. Moreover, surface tunable properties of these proteins help to bind a range of anticancer drugs and target ligands, thus making them efficient delivery agents in cancer therapy. Here, we discuss various proteins obtained from common exogenous sources and how they transform into effective anticancer agents. We also comprehensively discuss the tumor-killing mechanisms of different dietary proteins such as bovine α-lactalbumin, hen egg-white lysozyme, and their conjugates. We also articulate how protein nanostructures can be used as carriers for delivering cancer drugs and theranostics, and strategies to be adopted for improving their in vivo delivery and targeting. We further discuss the FDA-approved protein-based anticancer formulations along with those in different phases of clinical trials.
Collapse
|
19
|
Yamashita M, Kawakami N, Miyamoto K. Hydrophobization of a TIP60 Protein Nanocage for the Encapsulation of Hydrophobic Compounds. Chempluschem 2023; 88:e202200392. [PMID: 36775805 DOI: 10.1002/cplu.202200392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/14/2023]
Abstract
Encapsulation of hydrophobic molecules in protein-based nanocages is a promising approach for dispersing these molecules in water. Here, we report a chemical modification approach to produce a protein nanocage with a hydrophobic interior surface based on our previously developed nanocage, TIP60. The large pores of TIP60 act as tunnels for small molecules, allowing modification of the interior surface by hydrophobic compounds without nanocage disassembly. We used four different hydrophobic compounds for modification. The largest modification group tested, pyrene, resulted in a modified TIP60 that could encapsulate aromatic photosensitizer zinc phthalocyanine (ZnPC) more efficiently than the other modification compounds. The encapsulated ZnPC generated singlet oxygen upon light activation in the aqueous phase, whereas ZnPC alone formed inert aggregates under the same experimental conditions. Given that chemical modification allows a wider diversity of modifications than mutagenesis, this approach could be used to develop more suitable nanocages for encapsulating hydrophobic molecules of interest.
Collapse
Affiliation(s)
- Maika Yamashita
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223- 8522, Japan
| | - Norifumi Kawakami
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223- 8522, Japan
| | - Kenji Miyamoto
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223- 8522, Japan
| |
Collapse
|
20
|
Lu H, Cheng Z, Hu Y, Tang LV. What Can De Novo Protein Design Bring to the Treatment of Hematological Disorders? BIOLOGY 2023; 12:166. [PMID: 36829445 PMCID: PMC9952452 DOI: 10.3390/biology12020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Protein therapeutics have been widely used to treat hematological disorders. With the advent of de novo protein design, protein therapeutics are not limited to ameliorating natural proteins but also produce novel protein sequences, folds, and functions with shapes and functions customized to bind to the therapeutic targets. De novo protein techniques have been widely used biomedically to design novel diagnostic and therapeutic drugs, novel vaccines, and novel biological materials. In addition, de novo protein design has provided new options for treating hematological disorders. Scientists have designed protein switches called Colocalization-dependent Latching Orthogonal Cage-Key pRoteins (Co-LOCKR) that perform computations on the surface of cells. De novo designed molecules exhibit a better capacity than the currently available tyrosine kinase inhibitors in chronic myeloid leukemia therapy. De novo designed protein neoleukin-2/15 enhances chimeric antigen receptor T-cell activity. This new technique has great biomedical potential, especially in exploring new treatment methods for hematological disorders. This review discusses the development of de novo protein design and its biological applications, with emphasis on the treatment of hematological disorders.
Collapse
Affiliation(s)
| | | | | | - Liang V. Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
21
|
Kobayashi N, Arai R. Protein Cages and Nanostructures Constructed from Protein Nanobuilding Blocks. Methods Mol Biol 2023; 2671:79-94. [PMID: 37308639 DOI: 10.1007/978-1-0716-3222-2_4] [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: 06/14/2023]
Abstract
Protein cages and nanostructures are promising biocompatible medical materials, such as vaccines and drug carriers. Recent advances in designed protein nanocages and nanostructures have opened up cutting-edge applications in the fields of synthetic biology and biopharmaceuticals. A simple approach for constructing self-assembling protein nanocages and nanostructures is the design of a fusion protein composed of two different proteins forming symmetric oligomers. In this chapter, we describe the design and methods of protein nanobuilding blocks (PN-Blocks) using a dimeric de novo protein WA20 to construct self-assembling protein cages and nanostructures. A protein nanobuilding block (PN-Block), WA20-foldon, was developed by fusing an intermolecularly folded dimeric de novo protein WA20 and a trimeric foldon domain from bacteriophage T4 fibritin. The WA20-foldon self-assembled into several oligomeric nanoarchitectures in multiples of 6-mer. De novo extender protein nanobuilding blocks (ePN-Blocks) were also developed by fusing tandemly two WA20 with various linkers, to construct self-assembling cyclized and extended chain-like nanostructures. These PN-Blocks would be useful for the construction of self-assembling protein cages and nanostructures and their potential applications in the future.
Collapse
Affiliation(s)
- Naoya Kobayashi
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | - Ryoichi Arai
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Ueda, Nagano, Japan.
- Department of Applied Biology, Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano, Japan.
| |
Collapse
|
22
|
Kapelner RA, Fisher RS, Elbaum-Garfinkle S, Obermeyer AC. Protein charge parameters that influence stability and cellular internalization of polyelectrolyte complex micelles. Chem Sci 2022; 13:14346-14356. [PMID: 36545145 PMCID: PMC9749388 DOI: 10.1039/d2sc00192f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Proteins are an important class of biologics, but there are several recurring challenges to address when designing protein-based therapeutics. These challenges include: the propensity of proteins to aggregate during formulation, relatively low loading in traditional hydrophobic delivery vehicles, and inefficient cellular uptake. This last criterion is particularly challenging for anionic proteins as they cannot cross the anionic plasma membrane. Here we investigated the complex coacervation of anionic proteins with a block copolymer of opposite charge to form polyelectrolyte complex (PEC) micelles for use as a protein delivery vehicle. Using genetically modified variants of the model protein green fluorescent protein (GFP), we evaluated the role of protein charge and charge localization in the formation and stability of PEC micelles. A neutral-cationic block copolymer, poly(oligoethylene glycol methacrylate-block-quaternized 4-vinylpyridine), POEGMA79-b-qP4VP175, was prepared via RAFT polymerization for complexation and microphase separation with the panel of engineered anionic GFPs. We found that isotropically supercharged proteins formed micelles at higher ionic strength relative to protein variants with charge localized to a polypeptide tag. We then studied GFP delivery by PEC micelles and found that they effectively delivered the protein cargo to mammalian cells. However, cellular delivery varied as a function of protein charge and charge distribution and we found an inverse relationship between the PEC micelle critical salt concentration and delivery efficiency. This model system has highlighted the potential of polyelectrolyte complexes to deliver anionic proteins intracellularly. Using this model system, we have identified requirements for the formation of PEC micelles that are stable at physiological ionic strength and that smaller protein-polyelectrolyte complexes effectively deliver proteins to Jurkat cells.
Collapse
Affiliation(s)
- Rachel A Kapelner
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
| | - Rachel S Fisher
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
- Structural Biology Initiative, CUNY Advanced Science Research Center New York NY USA
| | - Shana Elbaum-Garfinkle
- Structural Biology Initiative, CUNY Advanced Science Research Center New York NY USA
- PhD Programs in Biochemistry and Biology at the Graduate Center, City University of New York NY USA
| | - Allie C Obermeyer
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
| |
Collapse
|
23
|
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: 7.5] [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
|
24
|
Gastrointestinal Tract Stabilized Protein Delivery Using Disulfide Thermostable Exoshell System. Int J Mol Sci 2022; 23:ijms23179856. [PMID: 36077259 PMCID: PMC9456531 DOI: 10.3390/ijms23179856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022] Open
Abstract
Thermostable exoshells (tES) are engineered proteinaceous nanoparticles used for the rapid encapsulation of therapeutic proteins/enzymes, whereby the nanoplatform protects the payload from proteases and other denaturants. Given the significance of oral delivery as the preferred model for drug administration, we structurally improved the stability of tES through multiple inter-subunit disulfide linkages that were initially absent in the parent molecule. The disulfide-linked tES, as compared to tES, significantly stabilized the activity of encapsulated horseradish peroxidase (HRP) at acidic pH and against the primary human digestive enzymes, pepsin, and trypsin. Furthermore, the disulfide-linked tES (DS-tES) exhibited significant intestinal permeability as evaluated using Caco2 cells. In vivo bioluminescence assay showed that encapsulated Renilla luciferase (rluc) was ~3 times more stable in mice compared to the free enzyme. DS-tES collected mice feces had ~100 times more active enzyme in comparison to the control (free enzyme) after 24 h of oral administration, demonstrating strong intestinal stability. Taken together, the in vitro and in vivo results demonstrate the potential of DS-tES for intraluminal and systemic oral drug delivery applications.
Collapse
|
25
|
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
|
26
|
Panjwani D, Patel S, Mishra D, Patel V, Yadav M, Dharamsi A, Patel A. Avidin-Biotin functionalized self-assembled protein nanoparticles as EGFR targeted therapeutics for the treatment of lung cancer: characterization and cell viability. J DISPER SCI TECHNOL 2022. [DOI: 10.1080/01932691.2022.2099888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Drishti Panjwani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Shruti Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Deepak Mishra
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Viral Patel
- Department of Civil and Petroleum Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - MangeRam Yadav
- Centre for Research and Development, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Abhay Dharamsi
- Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Asha Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
27
|
Weeks WB, Tainter CJ, Buchanan LE. Investigating the effects of N-terminal acetylation on KFE8 self-assembly with 2D IR spectroscopy. Biophys J 2022; 121:1549-1559. [PMID: 35247339 PMCID: PMC9072574 DOI: 10.1016/j.bpj.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/13/2021] [Accepted: 03/01/2022] [Indexed: 12/01/2022] Open
Abstract
Peptide self-assembly is an exciting and robust approach to create novel nanoscale materials for biomedical applications. However, the complex interplay between intra- and intermolecular interactions in peptide aggregation means that minor changes in peptide sequence can yield dramatic changes in supramolecular structure. Here, we use two-dimensional infrared (2D IR) spectroscopy to study a model amphiphilic peptide, KFE8, and its N-terminal acetylated counterpart, AcKFE8. 2D IR spectra of isotope-labeled peptides reveal that AcKFE8 aggregates comprise two distinct β-sheet structures while KFE8 aggregates comprise only one of these structures. Using an excitonic Hamiltonian to simulate the vibrational spectra of model β-sheets, we determine that the spectra are consistent with antiparallel β-sheets with different strand alignments, specifically a two-residue shift in the register of the β-strands. These findings bring forth new insights into how N-terminal acetylation may subtly impact secondary structure, leading to larger effects on overall aggregate morphology. Additionally, these results highlight the importance of understanding the residue-level structural differences that result from changes in peptide sequence in order to facilitate the rational design of peptide materials.
Collapse
Affiliation(s)
- William B Weeks
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Craig J Tainter
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | | |
Collapse
|
28
|
Habibi N, Mauser A, Ko Y, Lahann J. Protein Nanoparticles: Uniting the Power of Proteins with Engineering Design Approaches. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104012. [PMID: 35077010 PMCID: PMC8922121 DOI: 10.1002/advs.202104012] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/12/2021] [Indexed: 05/16/2023]
Abstract
Protein nanoparticles, PNPs, have played a long-standing role in food and industrial applications. More recently, their potential in nanomedicine has been more widely pursued. This review summarizes recent trends related to the preparation, application, and chemical construction of nanoparticles that use proteins as major building blocks. A particular focus has been given to emerging trends related to applications in nanomedicine, an area of research where PNPs are poised for major breakthroughs as drug delivery carriers, particle-based therapeutics or for non-viral gene therapy.
Collapse
Affiliation(s)
- Nahal Habibi
- Biointerfaces InstituteDepartment of Chemical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Ava Mauser
- Biointerfaces InstituteDepartment of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Yeongun Ko
- Biointerfaces InstituteDepartment of Chemical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Joerg Lahann
- Biointerfaces InstituteDepartments of Chemical EngineeringMaterial Science and EngineeringBiomedical Engineeringand Macromolecular Science and EngineeringUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
29
|
Miao Y, Yang T, Yang S, Yang M, Mao C. Protein nanoparticles directed cancer imaging and therapy. NANO CONVERGENCE 2022; 9:2. [PMID: 34997888 PMCID: PMC8742799 DOI: 10.1186/s40580-021-00293-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/29/2021] [Indexed: 05/10/2023]
Abstract
Cancer has been a serious threat to human health. Among drug delivery carriers, protein nanoparticles are unique because of their mild and environmentally friendly preparation methods. They also inherit desired characteristics from natural proteins, such as biocompatibility and biodegradability. Therefore, they have solved some problems inherent to inorganic nanocarriers such as poor biocompatibility. Also, the surface groups and cavity of protein nanoparticles allow for easy surface modification and drug loading. Besides, protein nanoparticles can be combined with inorganic nanoparticles or contrast agents to form multifunctional theranostic platforms. This review introduces representative protein nanoparticles applicable in cancer theranostics, including virus-like particles, albumin nanoparticles, silk protein nanoparticles, and ferritin nanoparticles. It also describes the common methods for preparing them. It then critically analyzes the use of a variety of protein nanoparticles in improved cancer imaging and therapy.
Collapse
Affiliation(s)
- Yao Miao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
| | - Tao Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058, Zhejiang, China.
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK, 73019-5251, USA.
| |
Collapse
|
30
|
Pan Y, Tang W, Fan W, Zhang J, Chen X. Development of nanotechnology-mediated precision radiotherapy for anti-metastasis and radioprotection. Chem Soc Rev 2022; 51:9759-9830. [DOI: 10.1039/d1cs01145f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radiotherapy (RT), including external beam RT and internal radiation therapy, uses high-energy ionizing radiation to kill tumor cells.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Wei Tang
- Departments of Pharmacy and Diagnostic Radiology, Nanomedicine Translational Research Program, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
31
|
Hango CR, Davis HC, Uddin EA, Minter LM, Tew GN. Increased block copolymer length improves intracellular availability of protein cargo. Polym Chem 2022. [DOI: 10.1039/d2py00017b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amphiphilic protein transduction domain mimics (PTDMs) of various lengths were used for protein delivery in Jurkat T cells. Although longer PTDMs facilitated greater cargo internalization, shorter PTDMs yielded greater cargo activity.
Collapse
Affiliation(s)
- Christopher R. Hango
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
| | - Hazel C. Davis
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
| | - Esha A. Uddin
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
| | - Lisa M. Minter
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
| | - Gregory N. Tew
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Amherst, Massachusetts 01003, USA
| |
Collapse
|
32
|
Shin MD, Hochberg JD, Pokorski JK, Steinmetz NF. Bioconjugation of Active Ingredients to Plant Viral Nanoparticles Is Enhanced by Preincubation with a Pluronic F127 Polymer Scaffold. ACS APPLIED MATERIALS & INTERFACES 2021; 13:59618-59632. [PMID: 34890195 DOI: 10.1021/acsami.1c13183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Proteinaceous nanoparticles can be used to deliver large payloads of active ingredients, which is advantageous in medicine and agriculture. However, the conjugation of hydrophobic ligands to hydrophilic nanocarriers such as plant viral nanoparticles (plant VNPs) can result in aggregation by reducing overall solubility. Given the benefits of hydrophilic nanocarrier platforms for targeted delivery and multivalent ligand display, coupled with the versatility of hydrophobic drugs, contrast agents, and peptides, this is an issue that must be addressed to realize their full potential. Here, we report two preincubation strategies that use a Pluronic F127 polymer scaffold to prevent the aggregation of conjugated plant VNPs: a plant VNP-polymer precoat (COAT) and an active ingredient formulation combined with a plant VNP-polymer precoat (FORMCOAT). The broad applications of these modified conjugation strategies were highlighted by testing their compatibility with three types of bioconjugation chemistry: N-hydroxysuccinimide ester-amine coupling, maleimide-thiol coupling, and copper(I)-catalyzed azide-alkyne cycloaddition (click chemistry). The COAT and FORMCOAT strategies promoted efficient bioconjugation and prevented the aggregation that accompanies conventional bioconjugation methods, thus improving the stability, homogeneity, and translational potential of plant VNP conjugates in medicine and agriculture.
Collapse
Affiliation(s)
- Matthew D Shin
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
| | - Justin D Hochberg
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
| | - Jonathan K Pokorski
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Institute for Materials Discovery and Design, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Institute for Materials Discovery and Design, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Department of Bioengineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Department of Radiology, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
- Moores Cancer Center, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92039, United States
| |
Collapse
|
33
|
Shariatinia Z. Big family of nano- and microscale drug delivery systems ranging from inorganic materials to polymeric and stimuli-responsive carriers as well as drug-conjugates. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
Achilli E, Flores C, Temprana C, Alonso SDV, Radrizzani M, Grasselli M. Enhanced gold nanoparticle-tumor cell recognition by albumin multilayer coating. OPENNANO 2021. [DOI: 10.1016/j.onano.2021.100033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Su R, Zhang Y, Zhang J, Wang H, Luo Y, Chan HF, Tao Y, Chen Z, Li M. Nanomedicine to advance the treatment of bacteria-induced acute lung injury. J Mater Chem B 2021; 9:9100-9115. [PMID: 34672317 DOI: 10.1039/d1tb01770e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bacteria-induced acute lung injury (ALI) is associated with a high mortality rate due to the lack of an effective treatment. Patients often rely on supportive care such as low tidal volume ventilation to alleviate the symptoms. Nanomedicine has recently received much attention owing to its premium benefits of delivering drugs in a sustainable and controllable manner while minimizing the potential side effects. It can effectively improve the prognosis of bacteria-induced ALI through targeted delivery of drugs, regulation of multiple inflammatory pathways, and combating antibiotic resistance. Hence, in this review, we first discuss the pathogenesis of ALI and its potential therapeutics. In particular, the state-of-the-art nanomedicines for the treatment of bacteria-induced ALI are highlighted, including their administration routes, in vivo distribution, and clearance. Furthermore, the available bacteria-induced ALI animal models are also summarized. In the end, future perspectives of nanomedicine for ALI treatment are proposed.
Collapse
Affiliation(s)
- Ruonan Su
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yu Zhang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca 14853, USA
| | - Jiabin Zhang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haixia Wang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhuanggui Chen
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
36
|
Teunissen AJP, Burnett ME, Prévot G, Klein ED, Bivona D, Mulder WJM. Embracing nanomaterials' interactions with the innate immune system. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1719. [PMID: 33847441 PMCID: PMC8511354 DOI: 10.1002/wnan.1719] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 03/21/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapy has firmly established itself as a compelling avenue for treating disease. Although many clinically approved immunotherapeutics engage the adaptive immune system, therapeutically targeting the innate immune system remains much less explored. Nanomedicine offers a compelling opportunity for innate immune system engagement, as many nanomaterials inherently interact with myeloid cells (e.g., monocytes, macrophages, neutrophils, and dendritic cells) or can be functionalized to target their cell-surface receptors. Here, we provide a perspective on exploiting nanomaterials for innate immune system regulation. We focus on specific nanomaterial design parameters, including size, form, rigidity, charge, and surface decoration. Furthermore, we examine the potential of high-throughput screening and machine learning, while also providing recommendations for advancing the field. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Abraham J. P. Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marianne E. Burnett
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Geoffrey Prévot
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emma D. Klein
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daniel Bivona
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Willem J. M. Mulder
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
37
|
Obata J, Kawakami N, Tsutsumi A, Nasu E, Miyamoto K, Kikkawa M, Arai R. Icosahedral 60-meric porous structure of designed supramolecular protein nanoparticle TIP60. Chem Commun (Camb) 2021; 57:10226-10229. [PMID: 34523636 DOI: 10.1039/d1cc03114g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Supramolecular protein nanoparticles and nanocages have potential in a broad range of applications. Recently, we developed a uniform supramolecular protein nanoparticle, TIP60, symmmetrically self-assembled from fusion proteins of a pentameric Sm-like protein and a dimeric MyoX-coil domain. Herein, we report the icosahedral 60-meric structure of TIP60 solved using single-particle cryo-electron microscopy. Interestingly, the structure revealed 20 regular-triangle-like pores on the surface. TIP60 and its mutants have many modifiable sites on their exterior and interior surfaces. The TIP60 architecture will be useful in the development of biomedical and biochemical nanoparticles/nanocages for future applications.
Collapse
Affiliation(s)
- Junya Obata
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Ueda, Nagano 386-8567, Japan. .,Department of Applied Biology, Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano 386-8567, Japan
| | - Norifumi Kawakami
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Akihisa Tsutsumi
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Erika Nasu
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Kenji Miyamoto
- Department of Bioscience and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Masahide Kikkawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Ryoichi Arai
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Ueda, Nagano 386-8567, Japan. .,Department of Applied Biology, Faculty of Textile Science and Technology, Shinshu University, Ueda, Nagano 386-8567, Japan
| |
Collapse
|
38
|
Chiesa E, Greco A, Riva F, Dorati R, Conti B, Modena T, Genta I. Hyaluronic Acid-Based Nanoparticles for Protein Delivery: Systematic Examination of Microfluidic Production Conditions. Pharmaceutics 2021; 13:1565. [PMID: 34683858 PMCID: PMC8539066 DOI: 10.3390/pharmaceutics13101565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
Hyaluronic acid-based nanoparticles (HA NPs) can be used to deliver a protein cargo to cells overexpressing HA receptors such as CD44 since they combine the low toxicity of the carrier and the retention of the protein integrity with the receptor-mediated internalization. HA properties play a crucial but sometimes unclear role in managing the formation and stability of the meshwork, cell interactions, and ultimately the protein entrapment efficacy. Nowadays, microfluidic is an innovative technology that allows to overcome limits linked to the NPs production, guaranteeing reproducibility and control of individual batches. Taking advantage of this technique, in this research work, the role of HA weight average molecular weight (Mw) in NPs formation inside a microfluidic device has been specifically faced. Based on the relationship between polymer Mw and solution viscosity, a methodological approach has been proposed to ensure critical quality attributes (size of 200 nm, PDI ≤ 0.3) to NPs made by HA with different Mw (280, 540, 710 and 820 kDa). The feasibility of the protein encapsulation was demonstrated by using Myoglobin, as a model neutral protein, with an encapsulation efficiency always higher than 50%. Lastly, all NPs samples were successfully internalized by CD44-expressing cells.
Collapse
Affiliation(s)
- Enrica Chiesa
- Department of Surgery, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Antonietta Greco
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Federica Riva
- Department of Public Health, Experimental and Forensic Medicine, Histology and Embryology Unit, University of Pavia, 27100 Pavia, Italy;
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Tiziana Modena
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.G.); (R.D.); (B.C.); (T.M.)
| |
Collapse
|
39
|
Larsen JB, Taebnia N, Dolatshahi-Pirouz A, Eriksen AZ, Hjørringgaard C, Kristensen K, Larsen NW, Larsen NB, Marie R, Mündler AK, Parhamifar L, Urquhart AJ, Weller A, Mortensen KI, Flyvbjerg H, Andresen TL. Imaging therapeutic peptide transport across intestinal barriers. RSC Chem Biol 2021; 2:1115-1143. [PMID: 34458827 PMCID: PMC8341777 DOI: 10.1039/d1cb00024a] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Oral delivery is a highly preferred method for drug administration due to high patient compliance. However, oral administration is intrinsically challenging for pharmacologically interesting drug classes, in particular pharmaceutical peptides, due to the biological barriers associated with the gastrointestinal tract. In this review, we start by summarizing the pharmacological performance of several clinically relevant orally administrated therapeutic peptides, highlighting their low bioavailabilities. Thus, there is a strong need to increase the transport of peptide drugs across the intestinal barrier to realize future treatment needs and further development in the field. Currently, progress is hampered by a lack of understanding of transport mechanisms that govern intestinal absorption and transport of peptide drugs, including the effects of the permeability enhancers commonly used to mediate uptake. We describe how, for the past decades, mechanistic insights have predominantly been gained using functional assays with end-point read-out capabilities, which only allow indirect study of peptide transport mechanisms. We then focus on fluorescence imaging that, on the other hand, provides opportunities to directly visualize and thus follow peptide transport at high spatiotemporal resolution. Consequently, it may provide new and detailed mechanistic understanding of the interplay between the physicochemical properties of peptides and cellular processes; an interplay that determines the efficiency of transport. We review current methodology and state of the art in the field of fluorescence imaging to study intestinal barrier transport of peptides, and provide a comprehensive overview of the imaging-compatible in vitro, ex vivo, and in vivo platforms that currently are being developed to accelerate this emerging field of research.
Collapse
Affiliation(s)
- Jannik Bruun Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Nayere Taebnia
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Alireza Dolatshahi-Pirouz
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Anne Zebitz Eriksen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Claudia Hjørringgaard
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Kasper Kristensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Nanna Wichmann Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Niels Bent Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Rodolphe Marie
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Ann-Kathrin Mündler
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Ladan Parhamifar
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Andrew James Urquhart
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Arjen Weller
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Kim I Mortensen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Henrik Flyvbjerg
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| | - Thomas Lars Andresen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark DK-2800, Kgs. Lyngby Denmark
| |
Collapse
|
40
|
Delfi M, Sartorius R, Ashrafizadeh M, Sharifi E, Zhang Y, De Berardinis P, Zarrabi A, Varma RS, Tay FR, Smith BR, Makvandi P. Self-assembled peptide and protein nanostructures for anti-cancer therapy: Targeted delivery, stimuli-responsive devices and immunotherapy. NANO TODAY 2021; 38:101119. [PMID: 34267794 PMCID: PMC8276870 DOI: 10.1016/j.nantod.2021.101119] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Self-assembled peptides and proteins possess tremendous potential as targeted drug delivery systems and key applications of these well-defined nanostructures reside in anti-cancer therapy. Peptides and proteins can self-assemble into nanostructures of diverse sizes and shapes in response to changing environmental conditions such as pH, temperature, ionic strength, as well as host and guest molecular interactions; their countless benefits include good biocompatibility and high loading capacity for hydrophobic and hydrophilic drugs. These self-assembled nanomaterials can be adorned with functional moieties to specifically target tumor cells. Stimuli-responsive features can also be incorporated with respect to the tumor microenvironment. This review sheds light on the growing interest in self-assembled peptides and proteins and their burgeoning applications in cancer treatment and immunotherapy.
Collapse
Affiliation(s)
- Masoud Delfi
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia, Naples 80126, Italy
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Naples 80131, Italy
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736, Hamadan, Iran
- Institute for Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Naples 80125, Italy
| | - Yapei Zhang
- Department of Biomedical Engineering, Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
| | | | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Rajender S. Varma
- Regional Centre of Advanced Technologies and Materials, Palacký University Olomouc, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA 30912, USA
| | - Bryan Ronain Smith
- Department of Biomedical Engineering, Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
- Department of Radiology and the Molecular Imaging Program, Stanford University, Stanford, CA, 94305, USA
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| |
Collapse
|
41
|
Sustained Release Systems for Delivery of Therapeutic Peptide/Protein. Biomacromolecules 2021; 22:2299-2324. [PMID: 33957752 DOI: 10.1021/acs.biomac.1c00160] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peptide/protein therapeutics have been significantly applied in the clinical treatment of various diseases such as cancer, diabetes, etc. owing to their high biocompatibility, specificity, and therapeutic efficacy. However, due to their immunogenicity, instability stemming from its complex tertiary and quaternary structure, vulnerability to enzyme degradation, and rapid renal clearance, the clinical application of protein/peptide therapeutics is significantly confined. Though nanotechnology has been demonstrated to prevent enzyme degradation of the protein therapeutics and thus enhance the half-life, issues such as initial burst release and uncontrollable release kinetics are still unsolved. Moreover, the traditional administration method results in poor patient compliance, limiting the clinical application of protein/peptide therapeutics. Exploiting the sustained-release formulations for more controllable delivery of protein/peptide therapeutics to decrease the frequency of injection and enhance patient compliance is thus greatly meaningful. In this review, we comprehensively summarize the substantial advancements of protein/peptide sustained-release systems in the past decades. In addition, the advantages and disadvantages of all these sustained-release systems in clinical application together with their future challenges are also discussed in this review.
Collapse
|
42
|
Khramtsov P, Kalashnikova T, Bochkova M, Kropaneva M, Timganova V, Zamorina S, Rayev M. Measuring the concentration of protein nanoparticles synthesized by desolvation method: Comparison of Bradford assay, BCA assay, hydrolysis/UV spectroscopy and gravimetric analysis. Int J Pharm 2021; 599:120422. [PMID: 33647407 DOI: 10.1016/j.ijpharm.2021.120422] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/04/2021] [Accepted: 02/21/2021] [Indexed: 12/12/2022]
Abstract
The desolvation technique is one of the most popular methods for preparing protein nanoparticles for medicine, biotechnology, and food applications. We fabricated 11 batches of BSA nanoparticles and 2 batches of gelatin nanoparticles by desolvation method. BSA nanoparticles from 2 batches were cross-linked by heating at +70 °C for 2 h; other nanoparticles were stabilized by glutaraldehyde. We compared several analytical approaches to measuring their concentration: gravimetric analysis, bicinchoninic acid assay, Bradford assay, and alkaline hydrolysis combined with UV spectroscopy. We revealed that the cross-linking degree and method of cross-linking affect both Bradford and BCA assay. Direct measurement of protein concentration in the suspension of purified nanoparticles by dye-binding assays can lead to significant (up to 50-60%) underestimation of nanoparticle concentration. Quantification of non-desolvated protein (indirect method) is affected by the presence of small nanoparticles in supernatants and can be inaccurate when the yield of desolvation is low. The reaction of cross-linker with protein changes UV absorbance of the latter. Therefore pure protein solution is an inappropriate calibrator when applying UV spectroscopy for the determination of nanoparticle concentration. Our recommendation is to determine the concentration of protein nanoparticles by at least two different methods, including gravimetric analysis.
Collapse
Affiliation(s)
- Pavel Khramtsov
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia.
| | - Tatyana Kalashnikova
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Maria Bochkova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Maria Kropaneva
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Valeria Timganova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Svetlana Zamorina
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| | - Mikhail Rayev
- Department of Biology, Perm State University, 614068, 15 Bukirev str., Perm, Russia; Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences, 614081, 13 Golev str., Perm, Russia
| |
Collapse
|
43
|
Rilo-Alvarez H, Ledo AM, Vidal A, Garcia-Fuentes M. Delivery of transcription factors as modulators of cell differentiation. Drug Deliv Transl Res 2021; 11:426-444. [PMID: 33611769 DOI: 10.1007/s13346-021-00931-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 12/13/2022]
Abstract
Fundamental studies performed during the last decades have shown that cell fate is much more plastic than previously considered, and technologies for its manipulation are a keystone for many new tissue regeneration therapies. Transcription factors (TFs) are DNA-binding proteins that control gene expression, and they have critical roles in the control of cell fate and other cellular behavior. TF-based therapies have much medical potential, but their use as drugs depends on the development of suitable delivery technologies that can help them reach their action site inside of the cells. TFs can be used either as proteins or encoded in polynucleotides. When used in protein form, many TFs require to be associated to a cell-penetrating peptide or another transduction domain. As polynucleotides, they can be delivered either by viral carriers or by non-viral systems such as polyplexes and lipoplexes. TF-based therapies have extensively shown their potential to solve many tissue-engineering problems, including bone, cartilage and cardiac regeneration. Yet, their use has expanded beyond regenerative medicine to other prominent disease areas such as cancer therapy and immunomodulation. This review summarizes some of the delivery options for effective TF-based therapies and their current main applications.
Collapse
Affiliation(s)
- Héctor Rilo-Alvarez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Adriana M Ledo
- Respiratory Therapeutic Area, Novartis Institutes for BioMedical Research, Inc, 700 Main Street, Cambridge, MA, 02139, USA
| | - Anxo Vidal
- Department of Physiology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
44
|
Wang J, Wu J, Li Y, Wen J, Cai J, Tang T, Hu X, Xiang D. The Brief Analysis of Peptide-combined Nanoparticle: Nanomedicine's Unique Value. Curr Protein Pept Sci 2021; 21:334-343. [PMID: 32039679 DOI: 10.2174/1389203721666200210103841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 08/20/2019] [Accepted: 09/25/2019] [Indexed: 12/21/2022]
Abstract
Therapeutic peptides (TPs) are biological macromolecules which can act as neurotransmitters, hormones, ion channel ligands and growth factors. Undoubtedly, TPs are crucial in modern medicine. But low bio-stability and some special adverse reactions reduce their places to the application. With the development of nanotechnology, nanoparticles (NPs) in pharmaceutical science gained much attention. They can encapsulate the TPs into their membrane or shell. Therefore, they can protect the TPs against degradation and then increase the bioavailability, which was thought to be the biggest advantage of them. Additionally, targeting was also studied to improve the effect of TPs. However, there were some drawbacks of nano TPs like low loading efficiency and difficulty to manufacture. Nowadays, lots of studies focused on improving effect of TPs by preparing nanoparticles. In this review, we presented a brief analysis of peptide-combined nanoparticles. Their advantages and disadvantages were listed in terms of mechanism. And several examples of applications were summarized.
Collapse
Affiliation(s)
- Jiemin Wang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yongjiang Li
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jing Wen
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jiaxin Cai
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiongbin Hu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
45
|
Chen K, Chen X, Han X, Fu Y. A comparison study on the release kinetics and mechanism of bovine serum albumin and nanoencapsulated albumin from hydrogel networks. Int J Biol Macromol 2020; 163:1291-1300. [DOI: 10.1016/j.ijbiomac.2020.07.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022]
|
46
|
Pustulka SM, Ling K, Pish SL, Champion JA. Protein Nanoparticle Charge and Hydrophobicity Govern Protein Corona and Macrophage Uptake. ACS APPLIED MATERIALS & INTERFACES 2020; 12:48284-48295. [PMID: 33054178 DOI: 10.1021/acsami.0c12341] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Protein nanoparticles are biomaterials composed entirely of proteins, with the protein sequence and structure determining the nanoparticle physicochemical properties. Upon exposure to physiological or environmental fluids, it is likely that protein nanoparticles, like synthetic nanoparticles, will adsorb proteins and this protein corona will be dependent on the surface properties of the protein nanoparticles. As there is little understanding of this phenomenon for engineered protein nanoparticles, the purpose of this work was to create protein nanoparticles with variable surface hydrophobicity and surface charge and establish the effect of these properties on the mass and composition of the adsorbed corona, using the fetal bovine serum as a model physiological solution. Albumin, cationic albumin, and ovalbumin cross-linked nanoparticles were developed for this investigation and their adsorbed protein coronas were isolated and characterized by gel electrophoresis and nanoliquid chromatography mass spectrometry. Distinct trends in corona mass and composition were identified for protein nanoparticles based on surface charge and surface hydrophobicity. Proteomic analyses revealed unique protein corona patterns and identified distinct proteins that are known to affect nanoparticle clearance in vivo. Further, the protein corona influenced nanoparticle internalization in vitro in a macrophage cell line. Altogether, these results demonstrate the strong effect protein identity and properties have on the corona formed on nanoparticles made from that protein. This work builds the foundation for future study of protein coronas on the wide array of protein nanoparticles used in nanomedicine and environmental applications.
Collapse
Affiliation(s)
- Samantha M Pustulka
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia 30332, United States
| | - Kevin Ling
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia 30332, United States
| | - Stephanie L Pish
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia 30332, United States
| | - Julie A Champion
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia 30332, United States
| |
Collapse
|
47
|
Quevedo DF, Lentz CJ, Coll de Peña A, Hernandez Y, Habibi N, Miki R, Lahann J, Lapizco-Encinas BH. Electrokinetic characterization of synthetic protein nanoparticles. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:1556-1567. [PMID: 33134000 PMCID: PMC7590587 DOI: 10.3762/bjnano.11.138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/29/2020] [Indexed: 05/11/2023]
Abstract
The application of nanoparticle in medicine is promising for the treatment of a wide variety of diseases. However, the slow progress in the field has resulted in relatively few therapies being translated into the clinic. Anisotropic synthetic protein nanoparticles (ASPNPs) show potential as a next-generation drug-delivery technology, due to their biocompatibility, biodegradability, and functionality. Even though ASPNPs have the potential to be used in a variety of applications, such as in the treatment of glioblastoma, there is currently no high-throughput technology for the processing of these particles. Insulator-based electrokinetics employ microfluidics devices that rely on electrokinetic principles to manipulate micro- and nanoparticles. These miniaturized devices can selectively trap and enrich nanoparticles based on their material characteristics, and subsequently release them, which allows for particle sorting and processing. In this study, we use insulator-based electrokinetic (EK) microdevices to characterize ASPNPs. We found that anisotropy strongly influences electrokinetic particle behavior by comparing compositionally identical anisotropic and non-anisotropic SPNPs. Additionally, we were able to estimate the empirical electrokinetic equilibrium parameter (eE EEC) for all SPNPs. This particle-dependent parameter can allow for the design of various separation and purification processes. These results show how promising the insulator-based EK microdevices are for the analysis and purification of clinically relevant SPNPs.
Collapse
Affiliation(s)
- Daniel F Quevedo
- Biointerfaces Institute, University of Michigan - Ann Arbor, Ann Arbor MI, USA
- Biomedical Engineering, University of Michigan - Ann Arbor, Ann Arbor MI, USA
| | - Cody J Lentz
- Microscale Bioseparations Laboratory and Biomedical Engineering Department, Rochester Institute of Technology, Rochester NY, USA
| | - Adriana Coll de Peña
- Microscale Bioseparations Laboratory and Biomedical Engineering Department, Rochester Institute of Technology, Rochester NY, USA
| | - Yazmin Hernandez
- Biointerfaces Institute, University of Michigan - Ann Arbor, Ann Arbor MI, USA
- Biomedical Engineering, University of Michigan - Ann Arbor, Ann Arbor MI, USA
| | - Nahal Habibi
- Biointerfaces Institute, University of Michigan - Ann Arbor, Ann Arbor MI, USA
- Chemical Engineering, University of Michigan - Ann Arbor, Ann Arbor MI, USA
| | - Rikako Miki
- Biointerfaces Institute, University of Michigan - Ann Arbor, Ann Arbor MI, USA
- Biomedical Engineering, University of Michigan - Ann Arbor, Ann Arbor MI, USA
| | - Joerg Lahann
- Biointerfaces Institute, University of Michigan - Ann Arbor, Ann Arbor MI, USA
- Biomedical Engineering, University of Michigan - Ann Arbor, Ann Arbor MI, USA
- Chemical Engineering, University of Michigan - Ann Arbor, Ann Arbor MI, USA
| | - Blanca H Lapizco-Encinas
- Microscale Bioseparations Laboratory and Biomedical Engineering Department, Rochester Institute of Technology, Rochester NY, USA
| |
Collapse
|
48
|
Baral A, Bhangu SK, Cimino R, Pelin JNBD, Alves WA, Chattopadhyay S, Ashokkumar M, Cavalieri F. Sono-Assembly of the [Arg-Phe] 4 Octapeptide into Biofunctional Nanoparticles. NANOMATERIALS 2020; 10:nano10091772. [PMID: 32911613 PMCID: PMC7558974 DOI: 10.3390/nano10091772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 12/22/2022]
Abstract
High-frequency ultrasound treatment is found to be a one-pot green technique to produce peptide-based nanostructures by ultrasound assisted self-assembly of oligopeptides. [Arg-Phe]4 octapeptides, consisting of alternating arginine (Arg/R) and phenylalanine (Phe/F) sequences, were subjected to 430 kHz ultrasound in aqueous solution in the absence of any external agents, to form [RF]4 nanoparticles ([RF]4-NPs), ~220 nm in diameter. A comprehensive analysis of the obtained nanoparticles demonstrated that the aromatic moieties of the oligopeptides can undergo oxidative coupling to form multiple oligomeric species, which then self-assemble into well-defined fluorescent nanoparticles. [RF]4-NPs were functionalized with polyethylene glycol (PEGylated) to improve their colloidal stability. Unlike the parent peptide, the PEGylated [RF]4-NPs showed limited cytotoxicity towards MDA-MB-231 cells. Furthermore, the intracellular trafficking of PEGylated [RF]4-NPs was investigated after incubation with MDA-MB-231 cells to demonstrate their efficient endo-lysosomal escape. This work highlights that the combined use of ultrasonic technologies and peptides enables easy fabrication of nanoparticles, with potential application in drug delivery.
Collapse
Affiliation(s)
- Anshul Baral
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia;
| | | | - Rita Cimino
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Juliane N. B. D. Pelin
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo Andre 09210-580, Brazil; (J.N.B.D.P.); (W.A.A.)
| | - Wendel A. Alves
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo Andre 09210-580, Brazil; (J.N.B.D.P.); (W.A.A.)
| | | | - Muthupandian Ashokkumar
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia;
- Correspondence: (M.A.); (F.C.)
| | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, VIC 3000, Australia;
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Correspondence: (M.A.); (F.C.)
| |
Collapse
|
49
|
Georgilis E, Abdelghani M, Pille J, Aydinlioglu E, van Hest JC, Lecommandoux S, Garanger E. Nanoparticles based on natural, engineered or synthetic proteins and polypeptides for drug delivery applications. Int J Pharm 2020; 586:119537. [DOI: 10.1016/j.ijpharm.2020.119537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
|
50
|
Habibi N, Christau S, Ochyl LJ, Fan Z, Hassani Najafabadi A, Kuehnhammer M, Zhang M, Helgeson M, Klitzing R, Moon JJ, Lahann J. Engineered Ovalbumin Nanoparticles for Cancer Immunotherapy. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000100] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nahal Habibi
- Department of Chemical Engineering University of Michigan Ann Arbor MI 48109 USA
- Biointerfaces Institute University of Michigan Ann Arbor MI 48109 USA
| | - Stephanie Christau
- Department of Chemical Engineering University of Michigan Ann Arbor MI 48109 USA
- Biointerfaces Institute University of Michigan Ann Arbor MI 48109 USA
| | - Lukasz J. Ochyl
- Biointerfaces Institute University of Michigan Ann Arbor MI 48109 USA
- Department of Pharmaceutical Sciences University of Michigan Ann Arbor MI 48109 USA
| | - Zixing Fan
- Department of Chemical Engineering University of Michigan Ann Arbor MI 48109 USA
| | - Alireza Hassani Najafabadi
- Biointerfaces Institute University of Michigan Ann Arbor MI 48109 USA
- Department of Pharmaceutical Sciences University of Michigan Ann Arbor MI 48109 USA
| | | | - Mengwen Zhang
- Department of Chemical Engineering University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Matthew Helgeson
- Department of Chemical Engineering University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Regine Klitzing
- Department of Physics Technische Universitaet Darmstadt Darmstadt 64289 Germany
| | - James J. Moon
- Biointerfaces Institute University of Michigan Ann Arbor MI 48109 USA
- Department of Pharmaceutical Sciences University of Michigan Ann Arbor MI 48109 USA
| | - Joerg Lahann
- Department of Chemical Engineering University of Michigan Ann Arbor MI 48109 USA
- Biointerfaces Institute University of Michigan Ann Arbor MI 48109 USA
| |
Collapse
|