1
|
Debnath M, Malhotra M, Kulkarni A. Protein corona formation on supramolecular polymer nanoparticles causes differential endosomal sorting resulting in an attenuated NLRP3 inflammasome activation. Biomater Sci 2025. [PMID: 40244934 DOI: 10.1039/d5bm00244c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Upon introduction into biological environments, nanoparticles undergo the spontaneous formation of a dynamic protein corona, which continually evolves and significantly modifies their physicochemical properties and interactions with biological systems. This evolving protein corona can critically impact the nanoparticles' endocytic pathways and targeting efficiency, potentially altering their functional characteristics and obscuring their intended therapeutic effects. Despite considerable focus on the characterization of corona proteins and their impact on nanoparticle uptake, the intracellular processes and their effects on immunogenicity are not yet thoroughly understood. Supramolecular polymer nanoparticles (SNPs) with a highly hydrophobic core are recognized for triggering NLRP3 inflammasome activation, a key component of the innate immune system. Here, it is reported that the protein corona formation on SNPs exerts an inhibitory effect on the activation pathway of NLRP3 inflammasome. The protein corona impairs the intrinsic capacity of SNPs to induce lysosomal membrane rupture, thereby diminishing the cellular stress signals necessary for the formation of the NLRP3 inflammasome complex. Furthermore, the cells transport SNPs with an attached protein corona to recycling endosomes, where they are sorted and prepared for exocytosis. Conversely, nascent SNPs are primarily confined to late endosomes and lysosomes, leading to lysosomal rupture and inflammasome activation. This differential routing reflects the significant impact of the protein corona on the cellular handling and subsequent biological activity of nanoparticles. In summary, this study elucidates the fundamental role of the protein corona in shaping the intracellular disposition of nanoparticles, with implications for modulating their interactions with the immune system.
Collapse
Affiliation(s)
- Maharshi Debnath
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.
| | - Mehak Malhotra
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
2
|
Wu H, Weng R, Li J, Huang Z, Tie X, Li J, Chen K. Self-Assembling protein nanoparticle platform for multivalent antigen delivery in vaccine development. Int J Pharm 2025; 676:125597. [PMID: 40233885 DOI: 10.1016/j.ijpharm.2025.125597] [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: 01/26/2025] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025]
Abstract
Nanoparticle vaccines can efficiently and repeatedly display multivalent antigens, thereby improving the targeted delivery of antigens and inducing more durable immune responses, making them an important representative of novel vaccines. The global COVID-19 pandemic has accelerated the development of nanoparticle vaccines, offering a promising solution for the prevention and control of infectious diseases. Currently, the development of nanoparticle vaccines involves the use of various types of nanoparticles, including liposomes, polymers, inorganic materials, and emulsions. Protein nanoparticles candidate vaccines are attracting increasing attention because of their unique antigen presentation methods and self-assembly characteristics during their development, leading to a broad consensus on their promising future. Naturally self-assembling protein nanoparticles, such as ferritin, enhance antigen presentation, which aids in the activation of both humoral and cellular immune responses. This has led to significant advancements in the study of hepatitis B virus. Meanwhile, some synthetically engineered protein nanoparticles, such as mi3, and I53-50, can induce higher antibody titers through chemical conjugation with the SpyTag-SpyCatcher system, thereby providing better immunoprotection and showing promising prospects in the prevention of H1N1 and H3N2 influenza virus infections. This article reviews the unique advantages of protein nanoparticles as antigen delivery platforms, progress made in immunological design mechanisms, advances in the application of related adjuvants in preclinical and clinical trials, and the performance of commonly used computationally designed protein nanoparticles in preclinical trials, with a particular emphasis on the progress in the application of cationic nanoparticle vaccines. The aim is to provide future researchers with effective adjuvant strategies and high-quality selections for computationally designed protein nanoparticles, thereby promoting the clinical trial process of protein nanoparticles vaccines.
Collapse
Affiliation(s)
- Hao Wu
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China; Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Ruiqi Weng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jiaxuan Li
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Zhiwei Huang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Xiaotian Tie
- Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Jianhua Li
- Zhejiang Key Laboratory of Public Health Detection and Pathogenesis Research, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, PR China.
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China.
| |
Collapse
|
3
|
Yoo J, Kim Y, Back JH, Shin J, Bae PK, Park KM, Kim M, Seo YH, Bak Y, Heo YH, Heo J, Choi H, Kim Y, Lee S, Lee JE, Jeong S, Yang JK, Kim S. Surface-engineered nanobeads for regioselective antibody binding: A robust immunoassay platform leveraging catalytic signal amplification. Biosens Bioelectron 2025; 281:117463. [PMID: 40228457 DOI: 10.1016/j.bios.2025.117463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/03/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Regulating protein interactions and protein corona formation of nanomaterials is crucial for advancing nanomedicine, where surface engineering of nanomaterials plays a pivotal role in precise control over biological interactions. Here, we present a surface-engineered nanoparticle-based immunoassay platform using carboxyl-enriched polystyrene nanobeads (CEPS) with regioselectively controlled antibody-binding properties. Proteomic analysis and theoretical simulation revealed that CEPS has an enhanced Fc-specific binding affinity for immunoglobulins compared to conventional carboxylated polystyrene beads, with a higher surface carboxyl density critically mediating protein interactions. This regioselective antibody binding with unique Fc-specific affinity eliminates the need for complex surface modifications, streamlining the assay process and broadening the applicability across various immunoassay formats. Additionally, incorporating a palladium catalyst within CEPS enables solvent-triggered on-demand catalytic signal amplification using a leucodye substrate, providing a more stable alternative to enzyme-based methods while significantly enhancing detection sensitivity and stability. The platform demonstrated enhanced performance in detecting clinically relevant biomarkers, including C-reactive protein, interferon-gamma, and the receptor-binding domain of SARS-CoV2, achieving lower detection limits and faster response times compared to conventional enzyme-based ELISA systems. Notably, the CEPS-based assay retained catalytic activity for over 140 days at room temperature, underscoring its potential for reliable, long-term use in diverse diagnostic applications.
Collapse
Affiliation(s)
- Jounghyun Yoo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Youngsun Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Hyun Back
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jawon Shin
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Pan Kee Bae
- HGUARD Inc., Daejeon, 34054, Republic of Korea
| | - Kyung Mi Park
- BioNano Health Guard Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Myung Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Young Hun Seo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; Biosensor Group, Korea Institute of Science and Technology Europe, Campus E7.1, Saarbrücken, 66123, Germany
| | - Yecheol Bak
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Yoon Ho Heo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Jeongyun Heo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Honghwan Choi
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Yongju Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Sangyoup Lee
- Bionic Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Eun Lee
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Sohdam Jeong
- Department of Chemical Engineering, Dong-Eui University, Busan, 47340, Republic of Korea.
| | - Jin-Kyoung Yang
- Department of Chemical Engineering, Dong-Eui University, Busan, 47340, Republic of Korea.
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
4
|
Razavi ZS, Razavi FS, Alizadeh SS. Inorganic nanoparticles and blood-brain barrier modulation: Advancing targeted neurological therapies. Eur J Med Chem 2025; 287:117357. [PMID: 39947054 DOI: 10.1016/j.ejmech.2025.117357] [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: 12/09/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
The blood-brain barrier (BBB) is a protective barrier that complicates the treatment of neurological disorders. Pharmaceutical compounds encounter significant challenges in crossing the central nervous system (CNS). Nanoparticles (NPs) are promising candidates for treating neurological conditions as they help facilitate drug delivery. This review explores the diverse characteristics and mechanisms of inorganic NPs (INPs), including metal-based, ferric-oxide, and carbon-based nanoparticles, which facilitate their passage through the BBB. Emphasis is placed on the physicochemical properties of NPs such as size, shape, surface charge, and surface modifications and their role in enhancing drug delivery efficacy, reducing immune clearance, and improving BBB permeability. Specific synthesis approaches are demonstrated, with an emphasis on the influence of each one on NP property, biological activity and the capability of an NP for its intended application. As for the advances in the field, the review emphasizes those characterized the NP formulation and surface chemistry that conquered the BBB and tested the need for its alteration. Current findings indicate that NP therapy can in the future enable effective targeting of specific brain disorders and eventually evolve this drug delivery system, which would allow for lower doses with less side effects.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
| | - Fateme Sadat Razavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | |
Collapse
|
5
|
Delavari B, Bigdeli B, Khazeni S, Varamini P. Nanodiamond-Protein hybrid Nanoparticles: LHRH receptor targeted and co-delivery of doxorubicin and dasatinib for triple negative breast cancer therapy. Int J Pharm 2025; 675:125544. [PMID: 40187703 DOI: 10.1016/j.ijpharm.2025.125544] [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: 11/12/2024] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Triple Negative Breast Cancer (TNBC) is an aggressive type of breast cancer that is difficult to treat with conventional therapies. This study aimed to develop a novel therapeutic approach using a multifunctional protein-nanodiamond nanocomposite to co-deliver doxorubicin (DOX) and dasatinib (DAS) to cancer cells via luteinising hormone-releasing hormone receptors. Nanodiamonds help retain DOX in targeted cells, while α-lactalbumin efficiently encapsulates DAS, reducing side effects. We successfully formulated the nanocomposite with over 80 % drug loading efficiency for both drugs. The imine Schiff-base bond in the nanocomposite hydrolyzes in the acidic pH tumor environment, triggering approximately 65 % drug release after 72 h, compared to less than 20 % in neutral pH. In vitro studies showed enhanced uptake of DOX and DAS in TNBC cell lines, potentially overcoming drug resistance. The combined delivery showed enhanced synergistic cytotoxic effects in drug-resistant TNBC cell models. For example, in the MDA-MB-231 cell line, the IC50 of DOX dropped to 45.63 ng/ml, while in MDA-MB-468, DAS decreased to 35.85 ng/ml with nanoparticle therapy. In vivo experiments utilizing a TNBC mouse model demonstrated the therapeutic effectiveness of the nanocomposite, leading to a 55 % reduction in tumor growth and enhanced survival rates. All mice given the nanocomposite survived after 44 days, but most treated with the DOX/DAS mixture died by day 28. This research showcases multifunctional nanocomposites as targeted drug delivery systems for TNBC, improving drug uptake and cytotoxicity. This strategy presents a promising method for treating drug-resistant breast cancer, with potential clinical applications and synergy with other therapies.
Collapse
Affiliation(s)
- Behdad Delavari
- School of Biomedical Science, Faculty of Medicine and Health, University of New South Wales, 2052 NSW, Australia; School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia.
| | - Bahareh Bigdeli
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia.
| | - Sepideh Khazeni
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia.
| | - Pegah Varamini
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, 2006 NSW, Australia; Nano Institute, University of Sydney, 2006 NSW, Australia.
| |
Collapse
|
6
|
Faezi M, Motavalizadehkakhky A, Homayouni Tabrizi M, Dolatabadi S, Es-Haghi A. Zein-sodium caseinate-diosmin nanoparticles as a promising anti-cancer agent with targeted efficacy against A2780 cell line. Sci Rep 2025; 15:8762. [PMID: 40082691 PMCID: PMC11906759 DOI: 10.1038/s41598-025-93772-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
This research investigated the potential of zein-sodium caseinate-diosmin nanoparticles (ZCD-NPs) as an anti-cancer agent against the A2780 cell line. Dynamic light scattering (DLS) analysis showed that ZCD-NPs have an average size of 265.30 nm with a polydispersity index of 0.21, indicating good uniformity suitable for pharmaceutical applications. Fourier transform infrared spectroscopy (FTIR) confirmed the successful incorporation of diosmin into the NPs and highlighted the interactions between the components. Field emission scanning electron microscopy (FESEM) images showed spherical NPs with smooth surfaces, suggesting stability and high production quality. Encapsulation efficiency was remarkably high, at 93.45%. Cytotoxicity assays showed a dose-dependent effect of ZCD-NPs, with A2780 cells showing significant sensitivity compared to normal HDF cells, indicating selective targeting of cancer cells. Flow cytometry analysis confirmed that ZCD-NPs induced apoptosis and necrosis in A2780 cells, as evidenced by increased expression of apoptotic genes such as p53 and caspases 8 and 9. In addition, ZCD-NPs exhibited potent antioxidant activity, effectively scavenging free radicals. These results suggest that ZCD-NPs have promising properties for targeted cancer therapy and antioxidant applications, which warrant further exploration in clinical settings.
Collapse
Affiliation(s)
- Mahmoud Faezi
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | | | | | - Samaneh Dolatabadi
- Department of Microbiology, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Ali Es-Haghi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
7
|
Ezerskyte E, Butkiene G, Katelnikovas A, Klimkevicius V. Development of Biocompatible, UV and NIR Excitable Nanoparticles with Multiwavelength Emission and Enhanced Colloidal Stability. ACS MATERIALS AU 2025; 5:353-364. [PMID: 40093831 PMCID: PMC11907297 DOI: 10.1021/acsmaterialsau.4c00151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 03/19/2025]
Abstract
The development of functional nanoprobes for biomedical applications is highly important in the field of modern nanotechnology. Due to strict requirements, such as the ability to be excited using irradiation, which allows deep tissue penetration, nonblinking behavior, and good optical and colloidal stability, the choice of nanoparticles is limited, and their synthesis is challenging. Among all of the functional nanoprobes for biomedical purposes, upconverting nanoparticles, especially those with more complex architectures (e.g., core-shell or core-shell-shell), are the most promising candidates. This study demonstrates advanced synthetic routes for constructing biocompatible nanoprobes with tunable optical properties and colloidal stability. The core-shell-shell architecture of the nanoprobes allows excitation from at least four sources, such as 272 and 394 nm of near-ultraviolet (near-UV) irradiation and 980 and 808 nm near-infrared (NIR) lasers. Furthermore, Gd-matrix-based nanoprobes doped with lanthanide ions (Nd3+, Yb3+, Tm3+, and Eu3+) are known for their paramagnetic properties for magnetic resonance imaging (MRI) imaging as well as upconversion luminescence with diverse emission bands across the entire visible spectrum. This feature is highly desirable for photodynamic therapy applications, as the upconversion emission of the proposed nanoprobes could overlap with the absorption band of commonly used photosensitizers and could potentially result in an efficient energy transfer process and enhanced generation of reactive oxygen species or singlet oxygen.
Collapse
Affiliation(s)
- Egle Ezerskyte
- Institute
of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko 24, LT-03225 Vilnius, Lithuania
- Biomedical
Physics Laboratory, National Cancer Institute, Baublio 3b, LT-08406 Vilnius, Lithuania
| | - Greta Butkiene
- Biomedical
Physics Laboratory, National Cancer Institute, Baublio 3b, LT-08406 Vilnius, Lithuania
| | - Arturas Katelnikovas
- Institute
of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko 24, LT-03225 Vilnius, Lithuania
| | - Vaidas Klimkevicius
- Institute
of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko 24, LT-03225 Vilnius, Lithuania
- Biomedical
Physics Laboratory, National Cancer Institute, Baublio 3b, LT-08406 Vilnius, Lithuania
| |
Collapse
|
8
|
Hong X, Chen T, Liu Y, Li J, Huang D, Ye K, Liao W, Wang Y, Liu M, Luan P. Design, current states, and challenges of nanomaterials in anti-neuroinflammation: A perspective on Alzheimer's disease. Ageing Res Rev 2025; 105:102669. [PMID: 39864562 DOI: 10.1016/j.arr.2025.102669] [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: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disease, brings huge damage to the society, to the whole family and even to the patient himself. However, until now, the etiological factor of AD is still unknown and there is no effective treatment for it. Massive deposition of amyloid-beta peptide(Aβ) and hyperphosphorylation of Tau proteins are acknowledged pathological features of AD. Recent studies have revealed that neuroinflammation plays a pivotal role in the pathology of AD. With the rise of nanomaterials in the biomedical field, researchers are exploring how the unique properties of these materials can be leveraged to develop effective treatments for AD. This article has summarized the influence of neuroinflammation in AD, the design of nanoplatforms, and the current research status and inadequacy of nanomaterials in improving neuroinflammation in AD.
Collapse
Affiliation(s)
- Xinyang Hong
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yunyun Liu
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Neurology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jun Li
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Dongqing Huang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Kaiyu Ye
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Wanchen Liao
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Yulin Wang
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Mengling Liu
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China.
| | - Ping Luan
- Department of Alzheimer's Disease Clinical Research Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China; School of Basic Medical Sciences, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
9
|
Wang X, Cao Z, Su J, Ma Y, Zhang S, Shao Z, Ge X, Cheng X, Zhou Z. Preparation of sodium alginate and chitosan modified curcumin liposomes and study on the formation of protein corona. Int J Biol Macromol 2025; 293:139392. [PMID: 39746413 DOI: 10.1016/j.ijbiomac.2024.139392] [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: 09/28/2024] [Revised: 12/05/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Curcumin (CUR) is a polyphenolic compound extracted from plants with a wide range of pharmacological activities. However, the low stability and bioavailability limits its practical application. This work utilized the chitosan (CH) and sodium alginate (SA) to modify the surface of the liposome to improve the stability of curcumin. Studies on the adsorption of pepsin to the surface of liposomes and the formation of protein coronas (PCs) were also carried out to investigate the in vivo behavior of the sodium alginate and chitosan modified curcumin liposomes (SA-CH-LPs). The result shown that the average particle size of SA-CH-LPs was around 220.7 ± 1.68 nm. X-ray Diffractometer (XRD) and differential scanning calorimeter (DSC) confirmed the successful preparation of SA-CH-LPs and illustrated their crystalline characteristics. The cytotoxicity of SA-CH-LPs was determined by CCK-8 assay and the results showed that the cell viability was above 80 % at different concentrations. In vitro results showed that the bioaccessibility of SA-CH-LPs (87.9 %) was better than curcumin liposomes (LPs) (66.4 %), and could better resist to catabolic degradation in the gastrointestinal environment. This work could provide the feasibility for improving the stability and bioaccessibility of the curcumin liposome, as well as given the preliminary evidence for investigation of the interaction with enzyme during the in vivo digestion process for the further application in food.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Zhaoxin Cao
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Jingyi Su
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Yu Ma
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Siyu Zhang
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Zihan Shao
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing 210037, PR China
| | - Xuemei Ge
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing 210037, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China.
| | - Xiaoliang Cheng
- Xi'an Jiaotong University, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Zhiyong Zhou
- College of Medicine and Health Sciences, Three Gorges University, Yichang, China
| |
Collapse
|
10
|
Bresinsky M, Goepferich A. Control of biomedical nanoparticle distribution and drug release in vivo by complex particle design strategies. Eur J Pharm Biopharm 2025; 208:114634. [PMID: 39826847 DOI: 10.1016/j.ejpb.2025.114634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
The utilization of targeted nanoparticles as a selective drug delivery system is a powerful tool to increase the amount of active substance reaching the target site. This can increase therapeutic efficacy while reducing adverse drug effects. However, nanoparticles face several challenges: upon injection, the immediate adhesion of plasma proteins may mask targeting ligands, thereby diminishing the target cell selectivity. In addition, opsonization can lead to premature clearance and the widespread presence of receptors or enzymes limits the accuracy of target cell recognition. Nanoparticles may also suffer from endosomal entrapment, and controlled drug release can be hindered by premature burst release or insufficient particle retention at the target site. Various strategies have been developed to address these adverse events, such as the implementation of switchable particle properties, regulating the composition of the formed protein corona, or using click-chemistry based targeting approaches. This has resulted in increasingly complex particle designs, raising the question of whether this development actually improves the therapeutic efficacy in vivo. This review provides an overview of the challenges in targeted drug delivery and explores potential solutions described in the literature. Subsequently, appropriate strategies for the development of nanoparticular drug delivery concepts are discussed.
Collapse
Affiliation(s)
- Melanie Bresinsky
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg 93053 Regensburg, Bavaria, Germany.
| |
Collapse
|
11
|
Hammond J, Richards CJ, Ko Y, Jonker T, Åberg C, Roos WH, Lira RB. Membrane Fusion-Based Drug Delivery Liposomes Transiently Modify the Material Properties of Synthetic and Biological Membranes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408039. [PMID: 40007088 PMCID: PMC11947515 DOI: 10.1002/smll.202408039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Many drug targets are located in intracellular compartments of cells but they often remain inaccessible to standard imaging and therapeutic agents. To aid intracellular delivery, drug carrier nanoparticles have been used to overcome the barrier imposed by the plasma membrane. The carrier must entrap large amounts of cargo, efficiently and quickly deliver the cargo in the cytosol or other intracellular compartments, and must be inert; they should not induce cellular responses or alter the cell state in the course of delivery. This study demonstrates that cationic liposomes with high charge density efficiently fuse with synthetic membranes and the plasma membrane of living cells. Direct fusion efficiently delivers large amounts of cargo to cells and cell-like vesicles within seconds, bypassing slow and often inefficient internalization-based pathways. These effects depend on liposome charge density, concentration, and the helper lipid. However, fusion-mediated cargo delivery results in the incorporation of large amounts of foreign lipids, causing changes to the material properties of these membranes, namely modifications in membrane packing and fluidity, induction of membrane curvature, decrease in surface tension, and the formation of (short-lived) pores. Importantly, these effects are transient and liposome removal allows cells to recover their state prior to liposome interaction.
Collapse
Affiliation(s)
- Jayna Hammond
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Ceri J. Richards
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyRijksuniversiteit GroningenGroningenThe Netherlands
| | - YouBeen Ko
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Thijs Jonker
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Christoffer Åberg
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyRijksuniversiteit GroningenGroningenThe Netherlands
| | - Wouter H. Roos
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Rafael B. Lira
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
- Present address:
Department of BionanoscienceKavli Institute of NanoscienceDelft University of TechnologyDelftThe Netherlands
| |
Collapse
|
12
|
Udupi A, Shetty S, Aranjani JM, Kumar R, Bharati S. Anticancer therapeutic potential of multimodal targeting agent- "phosphorylated galactosylated chitosan coated magnetic nanoparticles" against N-nitrosodiethylamine-induced hepatocellular carcinoma. Drug Deliv Transl Res 2025; 15:1023-1042. [PMID: 38990437 PMCID: PMC11782354 DOI: 10.1007/s13346-024-01655-1] [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] [Accepted: 06/19/2024] [Indexed: 07/12/2024]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) are extensively used as carriers in targeted drug delivery and has several advantages in the field of magnetic hyperthermia, chemodynamic therapy and magnet assisted radionuclide therapy. The characteristics of SPIONs can be tailored to deliver drugs into tumor via "passive targeting" and they can also be coated with tissue-specific agents to enhance tumor uptake via "active targeting". In our earlier studies, we developed HCC specific targeting agent- "phosphorylated galactosylated chitosan"(PGC) for targeting asialoglycoprotein receptors. Considering their encouraging results, in this study we developed a multifunctional targeting system- "phosphorylated galactosylated chitosan-coated magnetic nanoparticles"(PGCMNPs) for targeting HCC. PGCMNPs were synthesized by co-precipitation method and characterized by DLS, XRD, TEM, VSM, elemental analysis and FT-IR spectroscopy. PGCMNPs were evaluated for in vitro antioxidant properties, uptake in HepG2 cells, biodistribution, in vivo toxicity and were also evaluated for anticancer therapeutic potential against NDEA-induced HCC in mice model in terms of tumor status, electrical properties, antioxidant defense status and apoptosis. The characterization studies confirmed successful formation of PGCMNPs with superparamagnetic properties. The internalization studies demonstrated (99-100)% uptake of PGCMNPs in HepG2 cells. These results were also supported by biodistribution studies in which increased iron content (296%) was noted inside the hepatocytes. Further, PGCMNPs exhibited no in vivo toxicity. The anticancer therapeutic potential was evident from observation that PGCMNPs treatment decreased tumor bearing animals (41.6%) and significantly (p ≤ 0.05) lowered tumor multiplicity. Overall, this study indicated that PGCMNPs with improved properties are efficiently taken-up by hepatoma cells and has therapeutic potential against HCC. Further, this agent can be tagged with 32P and hence can offer multimodal cancer treatment options via radiation ablation as well as magnetic hyperthermia.
Collapse
Affiliation(s)
- Anushree Udupi
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sachin Shetty
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jesil Mathew Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rajesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Jodhpur, 342005, Rajasthan, India
| | - Sanjay Bharati
- Department of Nuclear Medicine, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
13
|
Gambaro R, Chain CY, Scioli-Montoto S, Moreno A, Huck-Iriart C, Ruiz ME, Cisneros JS, Lamas DG, Tau J, Gehring S, Islan GA, Rodenak-Kladniew B. Phytoactive-Loaded Lipid Nanocarriers for Simvastatin Delivery: A Drug Repositioning Strategy Against Lung Cancer. Pharmaceutics 2025; 17:255. [PMID: 40006622 PMCID: PMC11858925 DOI: 10.3390/pharmaceutics17020255] [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: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Drug repurposing explores new applications for approved medications, such as simvastatin (SV), a lipid-lowering drug that has shown anticancer potential but is limited by solubility and side effects. This study aims to enhance SV delivery and efficacy against lung cancer cells using bioactive lipid nanoparticles formulated with plant-derived monoterpenes as both nanostructuring agents and anticancer molecules. Methods: Lipid nanoparticles were produced by ultrasonication and characterized for morphology, size, zeta potential, and polydispersity index (PDI). Monoterpenes (linalool-LN-, limonene, 1,8-cineole) or Crodamol® were used as liquid lipids. Encapsulation efficiency (EE), release profiles, stability, biocompatibility, protein adsorption, cytotoxicity, and anticancer effects were evaluated. Results: The nanoparticles exhibited high stability, size: 94.2 ± 0.9-144.0 ± 2.6 nm, PDI < 0.3, and zeta potential: -4.5 ± 0.7 to -16.3 ± 0.8 mV. Encapsulation of SV in all formulations enhanced cytotoxicity against A549 lung cancer cells, with NLC/LN/SV showing the highest activity and being chosen for further investigation. Sustained SV release over 72 h and EE > 95% was observed for NLC/LN/SV. SAXS/WAXS analysis revealed that LN altered the crystallographic structure of nanoparticles. NLC/LN/SV demonstrated excellent biocompatibility and developed a thin serum protein corona in vitro. Cellular studies showed efficient uptake by A549 cells, G0/G1 arrest, mitochondrial hyperpolarization, reactive oxygen species production, and enhanced cell death compared to free SV. NLC/LN/SV more effectively inhibited cancer cell migration than free SV. Conclusions: NLC/LN/SV represents a promising nanocarrier for SV repurposing, combining enhanced anticancer activity, biocompatibility, and sustained stability for potential lung cancer therapy.
Collapse
Affiliation(s)
- Rocío Gambaro
- Children’s Hospital, University Medical Center of the Johannes, Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (R.G.); (S.G.)
| | - Cecilia Y. Chain
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), La Plata 1900, Buenos Aires, Argentina; (C.Y.C.); (J.S.C.)
| | - Sebastian Scioli-Montoto
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Buenos Aires, Argentina; (S.S.-M.); (M.E.R.)
| | - Ailin Moreno
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, Facultad de Ciencias Médicas UNLP, La Plata 1900, Buenos Aires, Argentina; (A.M.); (J.T.)
| | - Cristián Huck-Iriart
- Instituto de Tecnologías Emergentes y Ciencias Aplicadas (ITECA), Universidad Nacional de San Martín (UNSAM)--Investigaciones Científicas y Tecnológicas (CONICET), Escuela de Ciencia y Tecnología (ECyT), Laboratorio de Cristalografía Aplicada (LCA), Campus Miguelete, San Martín 1650, Buenos Aires, Argentina; (C.H.-I.); (D.G.L.)
- ALBA Synchrotron Light Source, Carrer de la Llum 2–26, Cerdanyola del Vallès, 08290 Barcelona, Spain
| | - María Esperanza Ruiz
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Buenos Aires, Argentina; (S.S.-M.); (M.E.R.)
| | - José S. Cisneros
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), La Plata 1900, Buenos Aires, Argentina; (C.Y.C.); (J.S.C.)
| | - Diego G. Lamas
- Instituto de Tecnologías Emergentes y Ciencias Aplicadas (ITECA), Universidad Nacional de San Martín (UNSAM)--Investigaciones Científicas y Tecnológicas (CONICET), Escuela de Ciencia y Tecnología (ECyT), Laboratorio de Cristalografía Aplicada (LCA), Campus Miguelete, San Martín 1650, Buenos Aires, Argentina; (C.H.-I.); (D.G.L.)
| | - Julia Tau
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, Facultad de Ciencias Médicas UNLP, La Plata 1900, Buenos Aires, Argentina; (A.M.); (J.T.)
| | - Stephan Gehring
- Children’s Hospital, University Medical Center of the Johannes, Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (R.G.); (S.G.)
| | - Germán A. Islan
- Children’s Hospital, University Medical Center of the Johannes, Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (R.G.); (S.G.)
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), Laboratorio de Nanobiomateriales, Departamento de Química, Facultad de Ciencias Exactas, Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, La Plata 1900, Buenos Aires, Argentina
| | - Boris Rodenak-Kladniew
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, Facultad de Ciencias Médicas UNLP, La Plata 1900, Buenos Aires, Argentina; (A.M.); (J.T.)
| |
Collapse
|
14
|
Chen K, Jiang Z, Yu J, Fu R, Yin H. Interaction of zinc oxide nanoparticles and nanorods with immunoglobulin G and underlying effects on MCF-7 breast cancer cells. Int J Biol Macromol 2025; 305:140924. [PMID: 39938843 DOI: 10.1016/j.ijbiomac.2025.140924] [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: 11/19/2024] [Revised: 02/08/2025] [Accepted: 02/09/2025] [Indexed: 02/14/2025]
Abstract
Protein corona formation is significantly impacted by the physicochemical characteristics of nanomaterials. The interaction of zinc oxide nanoparticles (ZnO NPs) and nanorods (NRs) with human immunoglobulin G (IgG) was investigated. Additionally, the anticancer effects of ZnO NPs and ZnO NRs in the forms of pristine or combined with protein coronas were evaluated in MCF-7 breast cancer cells. The results demonstrated that following the interaction with IgG, ZnO NPs with a hydrodynamic diameter (nm), zeta potential (mV), and UV-visible maximum wavelength (λmax, nm) of 9.75 ± 1.77, -21.89 ± 1.02, and 368 ± 0.32, respectively, showed a significant increase in size and λmax while decreasing in zeta potential. After ZnO NRs [hydrodynamic diameter (nm) of 68.45 ± 5.49, zeta potential (mV) of -33.04 ± 4.87, and λmax (nm) of 371 ± 0.48] interacted with IgG, these effects varied differently than those of ZnO NPs with IgG. The higher ΔG° value of the ZnO NR-IgG complex relative to the ZnO NP-IgG complex suggested greater stability of the formed complex in the presence of NRs than NPs. Additionally, in silico investigations demonstrated that the predominance of electrostatic forces promoted interactions between IgG and ZnO NPs or ZnO NRs. When interacting with ZnO NRs, the IgG conformation was more disrupted relative to ZnO NPs. Cellular studies showed that ZnO NPs, ZnO NRs, ZnO NP-IgG corona, and ZnO NR-IgG corona had IC50 values (μg/mL) of 161, 114, 118, and 62 μg/mL, respectively, in MCF-7 breast cancer cells. Additionally, qPCR and LDH assays demonstrated that, of all the examined groups, the ZnO NR-IgG corona caused the highest amount of cytotoxicity in MCF-7 breast cancer cells by altering the expression of Bax and Bcl-2 mRNA and triggering membrane damage.
Collapse
Affiliation(s)
- Keren Chen
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 528405, China.
| | - Zhuqiao Jiang
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 528405, China
| | - Jingwei Yu
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 528405, China
| | - Rao Fu
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 528405, China
| | - Hui Yin
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 528405, China
| |
Collapse
|
15
|
Tenchov R, Hughes KJ, Ganesan M, Iyer KA, Ralhan K, Lotti Diaz LM, Bird RE, Ivanov JM, Zhou QA. Transforming Medicine: Cutting-Edge Applications of Nanoscale Materials in Drug Delivery. ACS NANO 2025; 19:4011-4038. [PMID: 39823199 PMCID: PMC11803921 DOI: 10.1021/acsnano.4c09566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Since their inception in the early 1960s, the development and use of nanoscale materials have progressed tremendously, and their roles in diverse fields ranging from human health to energy and electronics are undeniable. The application of nanotechnology inventions has revolutionized many aspects of everyday life including various medical applications and specifically drug delivery systems, maximizing the therapeutic efficacy of the contained drugs by means of bioavailability enhancement or minimization of adverse effects. In this review, we utilize the CAS Content Collection, a vast repository of scientific information extracted from journal and patent publications, to analyze trends in nanoscience research relevant to drug delivery in an effort to provide a comprehensive and detailed picture of the use of nanotechnology in this field. We examine the publication landscape in the area to provide insights into current knowledge advances and developments. We review the major classes of nanosized drug delivery systems, their delivery routes, and targeted diseases. We outline the most discussed concepts and assess the advantages of various nanocarriers. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding nanosized drug delivery systems, to outline challenges, and to evaluate growth opportunities. The merit of the review stems from the extensive, wide-ranging coverage of the most up-to-date scientific information, allowing unmatched breadth of landscape analysis and in-depth insights.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Kevin J. Hughes
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Kavita A. Iyer
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Leilani M. Lotti Diaz
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert E. Bird
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Julian M. Ivanov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
16
|
Wang Z, Chen J, Wang J, Xu M, Yang H, Yang H, Zhao C, Sun P, Ji H, Liu J, Shan J, Tian J, Li S, Yu D, Wang C, Yu X, Ding S, Xu W, Zhang Y, Leng X, R-Porter T. MSCs biomimetic ultrasonic phase change nanoparticles promotes cardiac functional recovery after acute myocardial infarction. Biomaterials 2025; 313:122775. [PMID: 39241549 DOI: 10.1016/j.biomaterials.2024.122775] [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: 04/17/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/09/2024]
Abstract
Acute Myocardial Infarction (AMI) has seen rising cases, particularly in younger people, leading to public health concerns. Standard treatments, like coronary artery recanalization, often don't fully repair the heart's microvasculature, risking heart failure. Advances show that Mesenchymal Stromal Cells (MSCs) transplantation improves cardiac function after AMI, but the harsh microenvironment post-AMI impacts cell survival and therapeutic results. MSCs aid heart repair via their membrane proteins and paracrine extracellular vesicles that carry microRNA-125b, which regulates multiple targets, preventing cardiomyocyte death, limiting fibroblast growth, and combating myocardial remodeling after AMI. This study introduces ultrasound-responsive phase-change bionic nanoparticles, leveraging MSCs' natural properties. These particles contain MSC membrane and microRNA-125b, with added macrophage membrane for stability. Using Ultrasound Targeted Microbubble Destruction (UTMD), this method targets the delivery of MSC membrane proteins and microRNA-125b to AMI's inflamed areas. This aims to enhance cardiac function recovery and provide precise, targeted AMI therapy.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China; Key Laboratories of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, 150086, China
| | - Jianfeng Chen
- Laboratory Animal Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jiaxu Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China; Key Laboratories of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, 150086, China
| | - Mingyuan Xu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China
| | - Haichao Yang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China; Key Laboratories of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, 150086, China
| | - Haobo Yang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China; Key Laboratories of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, 150086, China
| | - Chen Zhao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China; Key Laboratories of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, 150086, China
| | - Huan Ji
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jinhong Liu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China
| | - Jiaxin Shan
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China
| | - Shouqiang Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China
| | - Dandan Yu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Chao Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xinhong Yu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Shuo Ding
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Wenjun Xu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Ying Zhang
- Ultrasound Imaging Department of the Second Affiliated Hospital of Harbin Medical University, and Pharmacology Department of Pharmacy College of Harbin Medical University, Harbin, 150081, China.
| | - Xiaoping Leng
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China; Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province (International Cooperation), Harbin, 150086, China.
| | - Thomas R-Porter
- Department of Cardiology, University of Nebraska Medical Center, Omaha, NE, NE 68198, USA
| |
Collapse
|
17
|
Quan Z, Wang S, Xie H, Zhang J, Duan R, Li M, Zhang J. ROS Regulation in CNS Disorder Therapy: Unveiling the Dual Roles of Nanomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410031. [PMID: 39676433 DOI: 10.1002/smll.202410031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/01/2024] [Indexed: 12/17/2024]
Abstract
The treatment of brain diseases has always been the focus of attention. Due to the presence of the blood-brain barrier (BBB), most small molecule drugs are difficult to reach the brain, leading to undesirable therapeutic outcomes. Recently, nanomedicines that can cross the BBB and precisely target lesion sites have emerged as thrilling tools to enhance the early diagnosis and treat various intractable brain disorders. Extensive research has shown that reactive oxygen species (ROS) play a crucial role in the occurrence and progression of brain diseases, including brain tumors and neurodegenerative diseases (NDDs) such as Alzheimer's disease, Parkinson's disease, stroke, or traumatic brain injury, making ROS a potential therapeutic target. In this review, on the structure and function of BBB as well as the mechanisms are first elaborated through which nanomedicine traverses it. Then, recent studies on ROS production are summarized through photodynamic therapy (PDT), chemodynamic therapy (CDT), and sonodynamic therapy (SDT) for treating brain tumors, and ROS depletion for treating NDDs. This provides valuable guidance for the future design of ROS-targeted nanomedicines for brain disease treatment. The ongoing challenges and future perspectives in developing nanomedicine-based ROS management for brain diseases are also discussed and outlined.
Collapse
Affiliation(s)
- Zhengyang Quan
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Sa Wang
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Huanhuan Xie
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Jiayi Zhang
- International department, Beijing 101 Middle School, Beijing, 100091, P. R. China
| | - Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
| | - Menglin Li
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Jinfeng Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, P. R. China
| |
Collapse
|
18
|
Upadhyaya J, Singh IR, Pun B, Baishya HJ, Kumar S, Joshi SR, Mitra S. Therapeutic Advantages of Nanoparticle-Impregnated Lysozyme Conjugates toward Amyloid-β Fibrillation and Antimicrobial Activity. J Phys Chem B 2025; 129:1274-1288. [PMID: 39812393 DOI: 10.1021/acs.jpcb.4c07261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The interaction of protein with nanoparticles (NPs) of varying shape and/or size boosts our understanding on their bioreactivity and establishes a comprehensive database for use in medicine, diagnosis, and therapeutic applications. The present study explores the interaction between lysozyme (LYZ) and different NPs like graphene oxide (GO) and zinc oxide (ZnO) having various shapes (spherical, 's', and rod-shaped, 'r') and sizes, focusing on their binding dynamics and subsequent effects on both the protein fibrillation and antimicrobial properties. Typically, GO is considered a promising medium due to its apparent inhibition and prolonged lag phase for LYZ fibrillation. However, the present results showed that spherical ZnO NPs (sZnO) offer superior efficacy in modulating fibrillation with an extended lag time of about 158.70 h, further emphasizing the importance of detailed investigation on the nanomaterial characteristics and fibril formation kinetics beyond initial observations. The experimental findings further confirmed a strong correlation between the binding affinity of NPs to the native protein and their effective inhibition of protein denaturation, ultimately preventing fibril formation. Interestingly, the lysozyme nanoconjugates showed intriguing bactericidal effects, as confirmed through the agar plate assay and SEM imaging, over the native protein. Overall, this study shows that appropriate bionanomaterials can exhibit multifunctional properties, which paves the way for a deeper investigation of NP characteristics, ultimately benefiting a wide array of intriguing research.
Collapse
Affiliation(s)
- Jahnabi Upadhyaya
- Department of Chemistry, North-Eastern Hill University, Shillong 793 022, India
| | | | - Bishal Pun
- Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong 793022, India
| | - Hirak Jyoti Baishya
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Sugam Kumar
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - S R Joshi
- Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong 793022, India
| | - Sivaprasad Mitra
- Department of Chemistry, North-Eastern Hill University, Shillong 793 022, India
| |
Collapse
|
19
|
Joung H, Jang GJ, Jeong JY, Lim G, Han SY. Evaluating the In Situ Effects of Whole Protein Coronas on the Biosensing of Antibody-Immobilized Nanoparticles Using Two-Color Fluorescence Nanoparticle Tracking Analysis. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:220. [PMID: 39940196 PMCID: PMC11820540 DOI: 10.3390/nano15030220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
The formation of protein coronas around engineered nanoparticles (ENPs) in biological environments is critical in nanomedicine, as these coronas significantly influence the biological behavior of ENPs. Despite extensive research on protein coronas, understanding the in situ influence of whole (soft plus hard) protein coronas has remained challenging. In this study, we demonstrate a strategy to assess the in situ effects of whole coronas on the model biosensing of anti-IgG using IgG-conjugated gold nanoparticles (IgG-AuNPs) through fluorescence nanoparticle tracking analysis (F-NTA), which enables the selective tracking of fluorescent particles within complex media. In our approach, anti-IgG and IgG-AuNPs were labeled with distinct fluorescent dyes. The accordance in hydrodynamic diameter distributions observed at two different wavelengths verifies the successful capture of anti-IgG on the IgG-AuNPs. The counting of fluorescent anti-IgG within the size distribution allows for a quantitative assessment of biosensing efficiency. This method was applied to evaluate the effects of four protein coronas-human serum albumin, high-density lipoproteins, immunoglobulin G, and fibrinogen-as well as their mixture across varying incubation times and concentrations. The results suggest that the physical presence of whole protein coronas surrounding the IgG-AuNPs may assist the biosensing interaction in situ rather than screening it.
Collapse
Affiliation(s)
| | | | | | | | - Sang Yun Han
- Department of Chemistry, Gachon University, Seongnam 13120, Gyeonggi, Republic of Korea; (H.J.); (G.J.J.)
| |
Collapse
|
20
|
Li XL, Yan ZS, Ma YQ, Ding HM. Impact of Glycosylation of Apolipoprotein D on Its Interaction with Gold Nanoparticles: Insights from Molecular Dynamics Simulations. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4490-4501. [PMID: 39772418 DOI: 10.1021/acsami.4c16685] [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: 01/11/2025]
Abstract
Efficient delivery of nanoparticles (NPs) as carriers for biochemical substances is crucial in various biomedical applications. In this study, we systematically investigate the interactions between glycosylated and nonglycosylated forms of Apolipoprotein D (ApoD) with gold nanoparticles (AuNPs) functionalized with different polymer coatings, including polyethylene glycol (PEG) and zwitterionic polymers. Using all-atom molecular dynamics simulations, we demonstrate that glycosylation significantly enhances the adsorption behavior of ApoD on AuNP surfaces, with the extent of this enhancement being dependent on the type (especially the charge property) of the polymer coatings. Notably, while zwitterionic polymers exhibit strong resistance to protein adsorption in their nonglycosylated form, this antifouling capability is diminished when glycosylation is present. Further, our findings reveal that glycosylation not only strengthens the binding energy of proteins but also alters the hydration dynamics at the NP-protein interface. Overall, this study provides a deeper understanding of the role of glycosylation in modulating protein-nanoparticle interactions, which is essential for the design of more effective nanomaterials for precision medicine.
Collapse
Affiliation(s)
- Xiao-Lei Li
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Zeng-Shuai Yan
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yu-Qiang Ma
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
- Jiangsu Physical Science Research Center, Nanjing 210093, China
| | - Hong-Ming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| |
Collapse
|
21
|
Van der Sanden N, Paun RA, Yitayew MY, Boyadjian O, Tabrizian M. An investigation of the effect of the protein corona on the cellular uptake of nanoliposomes under flow conditions using quartz crystal microgravimetry with dissipation. NANOSCALE ADVANCES 2024; 7:169-184. [PMID: 39569329 PMCID: PMC11575535 DOI: 10.1039/d4na00783b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024]
Abstract
When nanoparticle delivery systems are immersed in biological fluids, a complex assembly of proteins forms on their surface, creating a protein corona. The protein corona alters the physicochemical properties, toxicity, biodistribution, cellular uptake, and immune response of the nanoparticles, and consequently, their therapeutic efficacy. Currently, there is a lack of in vitro methods to assess the effects of the protein corona on nanoparticle uptake under dynamic flow and assess their binding kinetics in real-time. Here, we introduce quartz crystal microbalance with dissipation (QCM-D) as an in vitro technique, capable of incorporating dynamic flow, to study the effect of the protein corona on the binding of nanoliposome (NLP) formulations to cell surfaces as a first step in their cellular uptake. The interactions of four NLP formulations (low PEGylated, high PEGylated, negatively charged and positively charged NLPs) with A375 melanoma and THP1 cell lines were assessed by QCM-D, before and after the formation of a protein corona. Through real-time recording of the frequency and dissipation shifts (Δf and ΔD, respectively), the QCM-D results provided strong evidence of the role of the protein corona in the cellular interaction of these NLP formulations, with a variation in their adsorption kinetics depending on their initial composition. NLP's attachment to the cell surface was the lowest for PEGylated NLPs (<5%), while the positively charged NLPs showed the highest cellular attachment (≈100%), regardless of the presence of the protein corona or cell type. The effect of the protein corona was more pronounced for the negatively charged NLPs, where a significant reduction in the NLP attachment was observed. To complement the QCM-D data on the NLP attachment and to determine whether the NLP attachment leads to cellular uptake, confocal microscopy and flow cytometry were used to confirm NLP uptake by A375 and THP1 cells. Proteomic analysis revealed a differential composition of the protein corona on the various NLPs with possible implications for their sequestration and cellular uptake. Collectively, the findings suggest that QCM-D can be an important tool to study the binding of NLP formulations or other nanoparticles with cell membranes under dynamic flow, which very often differs from nanoparticle uptake under static conditions.
Collapse
Affiliation(s)
- Nicholas Van der Sanden
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Radu A Paun
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Michael Y Yitayew
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Oscar Boyadjian
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University Duff Medical Building, 3775 University Street Montreal Quebec H3A 2B4 Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University Montreal Canada
| |
Collapse
|
22
|
Akdaşçi E, Eker F, Duman H, Singh P, Bechelany M, Karav S. Lactoferrin as a Versatile Agent in Nanoparticle Applications: From Therapeutics to Agriculture. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2018. [PMID: 39728554 PMCID: PMC11728633 DOI: 10.3390/nano14242018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
Nanoparticles (NPs) have emerged as a potent choice for various applications, from drug delivery to agricultural studies, serving as an alternative and promising methodology for future advancements. They have been widely explored in delivery systems, demonstrating immense promise and high efficiency for the delivery of numerous biomolecules such as proteins and anticancer agents, either solely or modified with other compounds to enhance their capabilities. In addition, the utilization of NPs extends to antimicrobial studies, where they are used to develop novel antibacterial, antifungal, and antiviral formulations with advanced characteristics. Lactoferrin (Lf) is a glycoprotein recognized for its significant multifunctional properties, such as antimicrobial, antioxidant, anti-inflammatory, anticancer, and neuroprotective effects. Its activity has a broad distribution in the human body, with Lf receptors present in multiple regions. Current research shows that Lf is utilized in NP technology as a surface material, encapsulated biomolecule, and even as an NP itself. Due to the abundance of Lf receptors in various regions, Lf can be employed as a surface material in NPs for targeted delivery strategies, particularly in crossing the BBB and targeting specific cancers. Furthermore, Lf can be synthesized in an NP structure, positioning it as a strong candidate in future NP-related applications. In this article, we explore the highlighted and underexplored areas of Lf applications in NPs research.
Collapse
Affiliation(s)
- Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| | - Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| | - Priyanka Singh
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Mikhael Bechelany
- Institut Européen des Membranes (IEM), UMR 5635, University Montpellier, École Nationale Supérieure de Chimie de Montpellier (ENSCM), Centre National de la Recherche Scientifique (CNRS), F-34095 Montpellier, France
- Functional Materials Group, Gulf University for Science and Technology (GUST), Masjid Al Aqsa Street, Mubarak Al-Abdullah 32093, Kuwait
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| |
Collapse
|
23
|
Jia M, Ren W, Wang M, Liu Y, Wang C, Zhang Z, Xu M, Ding N, Li C, Yang H. Surface saturation of drug-loaded hollow manganese dioxide nanoparticles with human serum albumin for treating rheumatoid arthritis. Drug Deliv 2024; 31:2380538. [PMID: 39044468 PMCID: PMC11271085 DOI: 10.1080/10717544.2024.2380538] [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: 09/20/2023] [Accepted: 07/03/2024] [Indexed: 07/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease accompanied by energy depletion and accumulation of reactive oxygen species (ROS). Inorganic nanoparticles (NPs) offer great promise for the treatment of RA because they mostly have functions beyond being drug carriers. However, conventional nanomaterials become coated with a protein corona (PC) or lose their cargo prematurely in vivo, reducing their therapeutic efficacy. To avoid these problems, we loaded methotrexate (MTX) into hollow structured manganese dioxide nanoparticles (H-MnO2 NPs), then coated them with a 'pseudo-corona' of human serum albumin (HSA) at physiological concentrations to obtain HSA-MnO2@MTX NPs. Efficacy of MTX, MnO2@MTX, and HSA-MnO2@MTX NPs was compared in vitro and in vivo. Compared to MnO2@MTX, HSA-coated NPs were taken up better by lipopolysaccharide-activated RAW264.7 and were more effective at lowering levels of pro-inflammatory cytokines and preventing ROS accumulation. HSA-MnO2@MTX NPs were also more efficient at blocking the proliferation and migration of fibroblast-like synoviocytes from rats with collagen-induced arthritis. In this rat model, HSA-MnO2@MTX NPs showed better biodistribution than other treatments, specifically targeting the ankle joint. Furthermore, HSA-MnO2@MTX NPs reduced swelling in the paw, regulated pro-inflammatory cytokine production, and limited cartilage degradation and signs of inflammation. These results establish the therapeutic potential of HSA-MnO2@MTX NPs against RA.
Collapse
Affiliation(s)
- Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
- Nanchong Institute for Food and Drug Control, Nanchong, China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Minrui Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Nianhui Ding
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Hong Yang
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
- The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Kihara S, Aljabbari A, Bērziņš K, Krog LS, Mota-Santiago P, Terry A, Kirby N, Whitten AE, Boyd BJ. The "gut" corona at the surface of nanoparticles is dependent on exposure to bile salts and phospholipids. J Colloid Interface Sci 2024; 680:797-807. [PMID: 39591792 DOI: 10.1016/j.jcis.2024.11.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/07/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024]
Abstract
HYPOTHESIS The formation of a biomolecular corona on nanoparticle surfaces significantly influences their biological behaviour, particularly in drug delivery applications. Despite the prevalence of ingestion of particles (e.g, during oral drug delivery), our understanding of corona formation within the gastrointestinal (GI) tract remains limited, especially for non-protein components. The hypothesis of this work is that the exposure of nanoparticles to bile components will form a "corona" structure and protein corona will represent proteomes different from the original bile fluid. Two major aspects of biomolecular corona formed in GI fluid (hereby termed "gut corona), which ultimately dictate the fate of particle-based carriers, include the composition and the surface structure of nanoparticle-corona complex. EXPERIMENTS The structure and composition of the biomolecular corona formed on model SiO2 nanoparticles within simulated and extracted bile fluids were determined using small-angle scattering, quantification assays, and liquid chromatography with tandem mass spectrometry (LC-MS/MS) techniques. FINDINGS The formation of raspberry-like structures was identified, with bile micelles adopting ellipsoidal shapes around the nanoparticles, as opposed to a surface covered with a uniform corona (i.e., core-shell structure). Assay quantification and proteomics experiments revealed a notable increase in the ratio of protein to bile salt within the corona compared to the original bile fluid. The composition of the proteome differed between the bovine bile and the protein corona with only 34 proteins associated with the nanoparticles from the top 100 identified in bovine bile. Despite the differences in protein types identified between bovine bile and gut corona, the proportions of protein between different functional classes, such as enzymes and structural proteins, show little variation. This work elucidates the intricate interactions between nanoparticles and gut molecules, offering insights crucial for designing nanoparticle formulations for optimized oral drug delivery and understanding nanoparticle behaviour within the GI tract.
Collapse
Affiliation(s)
- Shinji Kihara
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| | - Anas Aljabbari
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Kārlis Bērziņš
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Lasse S Krog
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | | | - Ann Terry
- MAX IV Laboratory, Lund University, Lund, Sweden
| | - Nigel Kirby
- Australian Synchrotron, ANSTO, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Andrew E Whitten
- Australian Centre for Neutron Scattering (ACNS), Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, NSW 2234, Australia
| | - Ben J Boyd
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
25
|
Chen C, Alfredo YY, Lee YY, Tan CP, Wang Y, Qiu C. Physicochemical and biological characterization of the lipid particles with bovine serum albumin corona. Int J Biol Macromol 2024; 281:136223. [PMID: 39366617 DOI: 10.1016/j.ijbiomac.2024.136223] [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: 06/06/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
Diacylglycerol-based nanoparticles are promising bioactive delivery systems. However, limited understanding of their interaction with biological entities restricts their clinical use. This study investigated the protein corona formed on medium and long chain diacylglycerol (MLCD)-based solid lipid nanoparticles (NPs) modified by Polyoxethylene stearate (PEG) and compared to glyceryl tristearate (TG) and cetyl palmitate (CP) nanoparticles. Bovine serum albumin (BSA) formed corona with MLCD NPs through hydrophobic interactions and hydrogen bonding, contributing to a decrease in α-helix, an increase in β-sheet and a change in the microenvironment of Tyr residues. Owing to higher lipid hydrophilicity, MLCD NPs showed a much lower affinity for BSA than TG and CP NPs, and the binding constant with BSA was increased for larger NPs. PEG modification and the protein corona reduced the uptake of NPs by macrophages but exerted little influence on B16 cell. Among the NPs with different lipid core, the MLCD NPs showed a lower macrophages cell uptake but higher B16 cell uptake, suggesting a longer circulation time in blood but higher cancer cell internalization. This work shed light on the interactions between MLCD NPs and proteins, which is significant for application as nanocarriers with improved biological efficacy.
Collapse
Affiliation(s)
- Canfeng Chen
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Ying Ye Alfredo
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Yee Ying Lee
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Chin Ping Tan
- Department of Food Technology, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43300, Selangor, Malaysia
| | - Yong Wang
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China.
| | - Chaoying Qiu
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
26
|
Yan X, An N, Zhang Z, Qiu Q, Yang D, Wei P, Zhang X, Qiu L, Guo J. Graphene Oxide Quantum Dots-Preactivated Dental Pulp Stem Cells/GelMA Facilitates Mitophagy-Regulated Bone Regeneration. Int J Nanomedicine 2024; 19:10107-10128. [PMID: 39381026 PMCID: PMC11460356 DOI: 10.2147/ijn.s480979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024] Open
Abstract
Background In bone tissue engineering (BTE), cell-laden scaffolds offer a promising strategy for repairing bone defects, particularly when host cell regeneration is insufficient due to age or disease. Exogenous stem cell-based BTE requires bioactive factors to activate these cells. Graphene oxide quantum dots (GOQDs), zero-dimensional derivatives of graphene oxide, have emerged as potential osteogenic nanomedicines. However, constructing biological scaffolds with GOQDs and elucidating their biological mechanisms remain critical challenges. Methods We utilized GOQDs with a particle size of 10 nm, characterized by a surface rich in C-O-H and C-O-C functional groups. We developed a gelatin methacryloyl (GelMA) hydrogel incorporated with GOQDs-treated dental pulp stem cells (DPSCs). These constructs were transplanted into rat calvarial bone defects to estimate the effectiveness of GOQDs-induced DPSCs in repairing bone defects while also investigating the molecular mechanism underlying GOQDs-induced osteogenesis in DPSCs. Results GOQDs at 5 μg/mL significantly enhanced the osteogenic differentiation of DPSCs without toxicity. The GOQDs-induced DPSCs showed active osteogenic potential in three-dimensional cell culture system. In vivo, transplantation of GOQDs-preactivated DPSCs/GelMA composite effectively facilitated calvarial bone regeneration. Mechanistically, GOQDs stimulated mitophagy flux through the phosphatase-and-tensin homolog-induced putative kinase 1 (PINK1)/Parkin E3 ubiquitin ligase (PRKN) pathway. Notably, inhibiting mitophagy with cyclosporin A prevented the osteogenic activity of GOQDs. Conclusion This research presents a well-designed bionic GOQDs/DPSCs/GelMA composite scaffold and demonstrated its ability to promote bone regeneration by enhancing mitophagy. These findings highlight the significant potential of this composite for application in BTE and underscore the crucial role of mitophagy in promoting the osteogenic differentiation of GOQDs-induced stem cells.
Collapse
Affiliation(s)
- Xiaoyuan Yan
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Na An
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Zeying Zhang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Qiujing Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Di Yang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Penggong Wei
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Xiyue Zhang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Lihong Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Jiajie Guo
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| |
Collapse
|
27
|
Laganà A, Visalli G, Facciolà A, Saija C, Bertuccio MP, Baluce B, Celesti C, Iannazzo D, Di Pietro A. Sterile inflammation induced by respirable micro and nano polystyrene particles in the pathogenesis of pulmonary diseases. Toxicol Res (Camb) 2024; 13:tfae138. [PMID: 39233846 PMCID: PMC11368663 DOI: 10.1093/toxres/tfae138] [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: 05/07/2024] [Revised: 07/24/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024] Open
Abstract
Sterile inflammation is involved in the lung pathogenesis induced by respirable particles, including micro- and nanoplastics. Their increasing amounts in the ambient and in indoor air pose a risk to human health. In two human cell lines (A549 and THP-1) we assessed the proinflammatory behavior of polystyrene nanoplastics (nPS) and microplastics (mPS) (Ø 0.1 and 1 μm). Reproducing environmental aging, in addition to virgin, the cells were exposed to oxidized nPS/mPS. To study the response of the monocytes to the inflammatory signal transmitted by the A549 through the release of soluble factors (e.g. alarmins and cytokines), THP-1 cells were also exposed to the supernatants of previously nPS/mPS-treated A549. After dynamic-light-scattering (DLS) analysis and protein measurements for the assessment of protein corona in nPS/mPS, real-time PCR and enzyme-linked-immunosorbent (ELISA) assays were performed in exposed cells. The pro-inflammatory effects of v- and ox-nPS/mPS were attested by the imbalance of the Bax/Bcl-2 ratio in A549, which was able to trigger the inflammatory cascade, inhibiting the immunologically silent apoptosis. The involvement of NFkB was confirmed by the overexpression of p65 after exposure to ox-nPS and v- and ox-mPS. The fast and higher levels of IL-1β, only in THP-1 cells, underlined the NLPR3 inflammasome activation.
Collapse
Affiliation(s)
- Antonio Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
- Istituto Clinico Polispecialistico C.O.T. Cure Ortopediche Traumatologiche s.p.a., Viale Italia, 98124 Messina, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Caterina Saija
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Maria Paola Bertuccio
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Barbara Baluce
- Department of Transfusion Medicine and Hematology and Lombardy Regional Rare Blood Bank, IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122 Milan, Italy
| | - Consuelo Celesti
- Department of Electronic Engineering, Industrial Chemistry and Engineering, University of Messina, Via Stagno d'Alcontres, 98125 Messina, Italy
| | - Daniela Iannazzo
- Department of Electronic Engineering, Industrial Chemistry and Engineering, University of Messina, Via Stagno d'Alcontres, 98125 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
28
|
Morshed N, Rennie C, Deng W, Collins-Praino L, Care A. Serum-derived protein coronas affect nanoparticle interactions with brain cells. NANOTECHNOLOGY 2024; 35:495101. [PMID: 39284320 DOI: 10.1088/1361-6528/ad7b40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/16/2024] [Indexed: 09/20/2024]
Abstract
Neuronanomedicine is an emerging field bridging the gap between neuromedicine and novel nanotherapeutics. Despite promise, clinical translation of neuronanomedicine remains elusive, possibly due to a dearth of information regarding the effect of the protein corona on these neuronanomedicines. The protein corona, a layer of proteins adsorbed to nanoparticles following exposure to biological fluids, ultimately determines the fate of nanoparticles in biological systems, dictating nanoparticle-cell interactions. To date, few studies have investigated the effect of the protein corona on interactions with brain-derived cells, an important consideration for the development of neuronanomedicines. Here, two polymeric nanoparticles, poly(lactic-co-glycolic acid) (PLGA) and PLGA-polyethylene glycol (PLGA-PEG), were used to obtain serum-derived protein coronas. Protein corona characterization and liquid chromatography mass spectrometry analysis revealed distinct differences in biophysical properties and protein composition. PLGA protein coronas contained high abundance of globins (60%) and apolipoproteins (21%), while PLGA-PEG protein coronas contained fewer globins (42%) and high abundance of protease inhibitors (28%). Corona coated PLGA nanoparticles were readily internalized into microglia and neuronal cells, but not into astrocytes. Internalization of nanoparticles was associated with pro-inflammatory cytokine release and decreased neuronal cell viability, however, viability was rescued in cells treated with corona coated nanoparticles. These results showcase the importance of the protein corona in mediating nanoparticle-cell interactions.
Collapse
Affiliation(s)
- Nabila Morshed
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Claire Rennie
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Gadigal Country, Sydney, NSW 2007, Australia
| | - Lyndsey Collins-Praino
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
- Biologics Innovation Facility, University of Technology Sydney, Gadigal Country, Sydney, NSW 2007, Australia
| |
Collapse
|
29
|
Guo Y, Zhong W, Peng C, Guo L. Topical Delivery of Dual Loaded Nano-Transfersomes Mediated Chemo-Photodynamic Therapy against Melanoma via Inducing Cell Cycle Arrest and Apoptosis. Int J Mol Sci 2024; 25:9611. [PMID: 39273560 PMCID: PMC11394987 DOI: 10.3390/ijms25179611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Melanoma is a malignant skin cancer associated with high mortality rates and drug resistance, posing a significant threat to human health. The combination of chemotherapy and photodynamic therapy (PDT) represents a promising strategy to enhance antitumor efficacy through synergistic anti-cancer effects. Topical delivery of chemotherapeutic drugs and photosensitizers (PS) offers a non-invasive and safe way to treat melanoma. However, the effectiveness of these treatments is often hindered by challenges such as limited skin permeability and instability of the PS. In this study, transfersomes (TFS) were designed to facilitate transdermal delivery of the chemotherapeutic drug 5-Fluorouracil (5-FU) and the PS Imperatorin (IMP) for combined chemo-photodynamic therapy for melanoma. The cytotoxic and phototoxic effects of TFS-mediated PDT (TFS-UVA) were investigated in A375 cells and nude mice. The study also demonstrated that TFS-UVA generated intracellular ROS, induced G2/ M phase cell cycle arrest, and promoted cell apoptosis. In conclusion, this study indicated that 5-FU/ IMP-TFS serves as an effective transdermal therapeutic strategy for chemo-PDT in treating melanoma.
Collapse
Affiliation(s)
- Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wenxiao Zhong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
30
|
Zheng Y, Luo S, Xu M, He Q, Xie J, Wu J, Huang Y. Transepithelial transport of nanoparticles in oral drug delivery: From the perspective of surface and holistic property modulation. Acta Pharm Sin B 2024; 14:3876-3900. [PMID: 39309496 PMCID: PMC11413706 DOI: 10.1016/j.apsb.2024.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 09/25/2024] Open
Abstract
Despite the promising prospects of nanoparticles in oral drug delivery, the process of oral administration involves a complex transportation pathway that includes cellular uptake, intracellular trafficking, and exocytosis by intestinal epithelial cells, which are necessary steps for nanoparticles to enter the bloodstream and exert therapeutic effects. Current researchers have identified several crucial factors that regulate the interaction between nanoparticles and intestinal epithelial cells, including surface properties such as ligand modification, surface charge, hydrophilicity/hydrophobicity, intestinal protein corona formation, as well as holistic properties like particle size, shape, and rigidity. Understanding these properties is essential for enhancing transepithelial transport efficiency and designing effective oral drug delivery systems. Therefore, this review provides a comprehensive overview of the surface and holistic properties that influence the transepithelial transport of nanoparticles, elucidating the underlying principles governing their impact on transepithelial transport. The review also outlines the chosen of parameters to be considered for the subsequent design of oral drug delivery systems.
Collapse
Affiliation(s)
- Yaxian Zheng
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Shiqin Luo
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Min Xu
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin He
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiang Xie
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiawei Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
31
|
Wu Z, Ding Y, Qin Z, Sun Z, Wang Z, Cao X. Hemostatic Dressing Immobilized with ε-poly-L-lysine and Alginate Coated Mesoporous Bioactive Glass Prevents Blood Permeation by Pseudo-Dewetting Behavior. Adv Healthc Mater 2024; 13:e2400958. [PMID: 38770831 DOI: 10.1002/adhm.202400958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/07/2024] [Indexed: 05/22/2024]
Abstract
The integration of hemostats with cotton fabrics is recognized as an effective approach to improve the hemostatic performance of dressings. However, concerns regarding the uncontrollable absorption of blood by hydrophilic dressings and the risk of distal thrombosis from shed hemostatic agents are increasingly scrutinized. To address these issues, this work develops an advanced dressing (AQG) with immobilized nano-scale mesoporous bioactive glass (MBG) to safely and durably augment hemostasis. The doubly immobilized MBGs, pre-coated with ε-poly-L-lysine and alginate, demonstrate less than 1% detachment after ultrasonic washing. Notably, this MBG layer significantly promotes the adhesion, aggregation, and activation of red blood cells and platelets, adhered five times more red blood cells and 29 times more platelets than raw dressing, respectively. Specially, with the rapid formation of protein corona and amplification of thrombin, dense fibrin network is built on MBG layer and then blocked blood permeation transversely and longitudinally, showing an autophobic pseudo-dewetting behavior and allowing AQG to concentrate blood in situ and culminate in faster hemostasis with lower blood loss. Furthermore, the potent antibacterial properties of AQG extend its potential for broader application in daily care and clinical setting.
Collapse
Affiliation(s)
- Zilin Wu
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Yilin Ding
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Zhihao Qin
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Zhipeng Sun
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Zetao Wang
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Xiaodong Cao
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| |
Collapse
|
32
|
Jain M, Yu X, Schneck JP, Green JJ. Nanoparticle Targeting Strategies for Lipid and Polymer-Based Gene Delivery to Immune Cells In Vivo. SMALL SCIENCE 2024; 4:2400248. [PMID: 40212067 PMCID: PMC11935263 DOI: 10.1002/smsc.202400248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/15/2024] [Indexed: 04/13/2025] Open
Abstract
Lipid nanoparticles and polymeric nanoparticles are promising biomaterial platforms for robust intracellular DNA and mRNA delivery, highlighted by the widespread use of nanoparticle- (NP) based mRNA vaccines to help end the COVID-19 pandemic. Recent research has sought to adapt this nanotechnology to transfect and engineer immune cells in vivo. The immune system is an especially appealing target due to its involvement in many different diseases, and ex vivo-engineered immune cell therapies like chimeric antigen receptor (CAR) T therapy have already demonstrated remarkable clinical success in certain blood cancers. Although gene delivery can potentially address some of the cost and manufacturing concerns associated with current autologous immune cell therapies, transfecting immune cells in vivo is challenging. Not only is extrahepatic NP delivery to lymphoid organs difficult, but immune cells like T cells have demonstrated particular resistance to transfection. Despite these challenges, the modular nature of NPs allows researchers to examine critical structure-function relationships between a particle's properties and its ability to specifically engineer immune cells in vivo. Herein, several nanomaterial components are outlined, including targeting ligands, nucleic acid cargo, chemical properties, physical properties, and the route of administration to specifically target NPs to immune cells for optimal in vivo transfection.
Collapse
Affiliation(s)
- Manav Jain
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Xinjie Yu
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jonathan P. Schneck
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Pathology and MedicineJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Jordan J. Green
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of OncologyThe Sidney Kimmel Comprehensive Cancer CenterThe Bloomberg∼Kimmel Institute for Cancer ImmunotherapyJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Ophthalmology, Neurosurgery, and Materials Science & EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
33
|
Kachanov A, Kostyusheva A, Brezgin S, Karandashov I, Ponomareva N, Tikhonov A, Lukashev A, Pokrovsky V, Zamyatnin AA, Parodi A, Chulanov V, Kostyushev D. The menace of severe adverse events and deaths associated with viral gene therapy and its potential solution. Med Res Rev 2024; 44:2112-2193. [PMID: 38549260 DOI: 10.1002/med.22036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, in vivo gene replacement therapy has significantly advanced, resulting in market approval of numerous therapeutics predominantly relying on adeno-associated viral vectors (AAV). While viral vectors have undeniably addressed several critical healthcare challenges, their clinical application has unveiled a range of limitations and safety concerns. This review highlights the emerging challenges in the field of gene therapy. At first, we discuss both the role of biological barriers in viral gene therapy with a focus on AAVs, and review current landscape of in vivo human gene therapy. We delineate advantages and disadvantages of AAVs as gene delivery vehicles, mostly from the safety perspective (hepatotoxicity, cardiotoxicity, neurotoxicity, inflammatory responses etc.), and outline the mechanisms of adverse events in response to AAV. Contribution of every aspect of AAV vectors (genomic structure, capsid proteins) and host responses to injected AAV is considered and substantiated by basic, translational and clinical studies. The updated evaluation of recent AAV clinical trials and current medical experience clearly shows the risks of AAVs that sometimes overshadow the hopes for curing a hereditary disease. At last, a set of established and new molecular and nanotechnology tools and approaches are provided as potential solutions for mitigating or eliminating side effects. The increasing number of severe adverse reactions and, sadly deaths, demands decisive actions to resolve the issue of immune responses and extremely high doses of viral vectors used for gene therapy. In response to these challenges, various strategies are under development, including approaches aimed at augmenting characteristics of viral vectors and others focused on creating secure and efficacious non-viral vectors. This comprehensive review offers an overarching perspective on the present state of gene therapy utilizing both viral and non-viral vectors.
Collapse
Affiliation(s)
- Artyom Kachanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Ivan Karandashov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Andrey Tikhonov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Vadim Pokrovsky
- Laboratory of Biochemical Fundamentals of Pharmacology and Cancer Models, Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People's Friendship University, Russia (RUDN University), Moscow, Russia
| | - Andrey A Zamyatnin
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Research, Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Vladimir Chulanov
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Infectious Diseases, Sechenov University, Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
34
|
Longobardi G, Moore TL, Conte C, Ungaro F, Satchi‐Fainaro R, Quaglia F. Polyester nanoparticles delivering chemotherapeutics: Learning from the past and looking to the future to enhance their clinical impact in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1990. [PMID: 39217459 PMCID: PMC11670051 DOI: 10.1002/wnan.1990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Polymeric nanoparticles (NPs), specifically those comprised of biodegradable and biocompatible polyesters, have been heralded as a game-changing drug delivery platform. In fact, poly(α-hydroxy acids) such as polylactide (PLA), poly(lactide-co-glycolide) (PLGA), and poly(ε-caprolactone) (PCL) have been heavily researched in the past three decades as the material basis of polymeric NPs for drug delivery applications. As materials, these polymers have found success in resorbable sutures, biodegradable implants, and even monolithic, biodegradable platforms for sustained release of therapeutics (e.g., proteins and small molecules) and diagnostics. Few fields have gained more attention in drug delivery through polymeric NPs than cancer therapy. However, the clinical translational of polymeric nanomedicines for treating solid tumors has not been congruent with the fervor or funding in this particular field of research. Here, we attempt to provide a comprehensive snapshot of polyester NPs in the context of chemotherapeutic delivery. This includes a preliminary exploration of the polymeric nanomedicine in the cancer research space. We examine the various processes for producing polyester NPs, including methods for surface-functionalization, and related challenges. After a detailed overview of the multiple factors involved with the delivery of NPs to solid tumors, the crosstalk between particle design and interactions with biological systems is discussed. Finally, we report state-of-the-art approaches toward effective delivery of NPs to tumors, aiming at identifying new research areas and re-evaluating the reasons why some research avenues have underdelivered. We hope our effort will contribute to a better understanding of the gap to fill and delineate the future research work needed to bring polyester-based NPs closer to clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Thomas Lee Moore
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Claudia Conte
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Francesca Ungaro
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Sagol School of NeurosciencesTel Aviv UniversityTel AvivIsrael
| | - Fabiana Quaglia
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
35
|
Yang Y, Chu Y, Li C, Fan L, Lu H, Zhan C, Zhang Z. Brain-targeted drug delivery by in vivo functionalized liposome with stable D-peptide ligand. J Control Release 2024; 373:240-251. [PMID: 38977135 DOI: 10.1016/j.jconrel.2024.07.014] [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: 04/09/2024] [Revised: 06/17/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Brain-targeted drug delivery poses a great challenge due to the blood-brain barrier (BBB). In a previous study, we have developed a peptide-modified stealth liposome (SP-sLip) to enhance BBB penetration via the adsorption of apolipoproteins in plasma. SP is an 11-amino acid peptide derived from 25 to 35 of the Amyloid β peptide (Aβ1-42), which is a nature ligand of apolipoproteins. Although freshly prepared SP-sLip exhibited efficient brain targeting performance, it occured self-aggregation and instability in storage. In this study, we developed a D-peptide ligand according to the reverse sequence of SP with D-amino acids, known as DSP, to improve the stability in storage. Notably, DSP exhibited a reduced tendency for self-aggregation and improved stability in comparison to the SP peptide. Furthermore, compared to SP-sLip, DSP-modified sLip (DSP-sLip) demonstrated enhanced stability (>2 weeks), prolonged blood circulation (AUC increased 44.4%), reduced liver and spleen accumulation (reduced by 2.23 times and 1.86 times) with comparable brain-targeting efficiency. Similar to SP-sLip, DSP-sLip selectively adsorbed apolipoprotein A1, E, and J in the blood to form functionalized protein corona, thus crossing BBB via apolipoprotein receptor-mediated transcytosis. These findings underscored the importance of ligand stability in the in vitro and in vivo performance of brain-targeted liposomes, therefore paving the way for the design and optimization of efficient and stable nanocarriers.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Yuxiu Chu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Cheng Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Lianfeng Fan
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Huiping Lu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China.
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China.
| | - Zui Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
36
|
Zhou J, Shen W, Feng W, Zhang X, Wu T, Zhou J, Su Z, Yin T. Temperature Self-Limited Intelligent Thermo-chemotherapeutic Lipid Nanosystem for P-gp Reversal Time Window Matched Pulse Treatment of MDR Tumor. NANO LETTERS 2024; 24:10631-10641. [PMID: 39150779 DOI: 10.1021/acs.nanolett.4c02978] [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: 08/18/2024]
Abstract
Mild photothermal therapy (PTT) shows the potential for chemosensitization by tumor-localized P-glycoprotein (P-gp) modulation. However, conventional mild PTT struggles with real-time uniform temperature control, obscuring the temperature-performance relationship and resulting in thermal damage. Besides, the time-performance relationship and the underlying mechanism of mild PTT-mediated P-gp reversal remains elusive. Herein, we developed a temperature self-limiting lipid nanosystem (RFE@PD) that integrated a reversible organic heat generator (metal-phenolic complexes) and metal chelator (deferiprone, DFP) encapsulated phase change material. Upon NIR irradiation, RFE@PD released DFP for blocking ligand-metal charge transfer to self-limit temperature below 45 °C, and rapidly reduced P-gp within 3 h via Ubiquitin-proteasome degradation. Consequently, the DOX·HCl-loaded thermo-chemotherapeutic lipid nanosystem (RFE@PD-DOX) led to dramatically improved drug accumulation and 5-fold chemosensitization in MCF-7/ADR tumor models by synchronizing P-gp reversal and drug pulse liberation, achieving a tumor inhibition ratio of 82.42%. This lipid nanosystem integrated with "intrinsic temperature-control" and "temperature-responsive pulse release" casts new light on MDR tumor therapy.
Collapse
Affiliation(s)
- Jiyuan Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Weiyang Shen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Wenna Feng
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xin Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Tongyu Wu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Zhigui Su
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| |
Collapse
|
37
|
Huang Y, Ye D, Liu X, Chen H, Luo X, Huang B, Zhou N, Wang H, Zou Q, Fang S, Wang S, Wu L. Spiky tubular nanoparticles with low protein corona can realize efficient and non-destructive penetration through endothelial barrier. J Control Release 2024; 374:1-14. [PMID: 39079657 DOI: 10.1016/j.jconrel.2024.07.060] [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: 02/10/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Upon intravascular applications, i.e., cancer treatment, nanoparticles (NPs) are required to deliver through blood circulation, sustain serum protein interactions, before they penetrate the blood vessels and reach targeted sites for payload drug release. For a delivery process as such, it is elusive and difficult to comprehend the morphological change of NP surface and evaluate associated effects on its targeted delivery. Herein, we used silica NPs with different surface modifications to demonstrate the morphological impact of NPs during the application of the NP-blood protein interaction, vascular endothelial cell penetration, subsequent targeted delivery and photodynamic therapy efficacy, and pursue high drug-load NPs with surface designs. Compared to solid and mesoporous NPs, we found the spiky tubular NPs reserved the NPs' antifouling properties (or shedding of "protein corona"), promoted better endothelial penetration and less destruction in vitro and in vivo. Such effects could be attributed to their spiky surface structures, which can limit the NP-protein interaction area and promote the NP-protein steric hindrance. Further in molecular simulations, we determined that the spiky tubular morphological modification on NPs enhanced the interaction free energy and lowered the amino acids number and the subsequent frequency in contacting with VE-cadherin of vascular endothelia. As a result, the spiky tubular NPs demonstrated its advantages in mitigating damages to VE-cadherin stability and endothelial cell integrity. Exploiting such spiky tubular surface modification, we can improve the NP delivery efficiency and prohibit the leakiness of vascular endothelia, helping address challenges faced by tumor migration in nanomedicine applications for cancer therapy.
Collapse
Affiliation(s)
- Yuan Huang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Dong Ye
- Molecular Profiling and Drug Delivery, Small Molecule CMC Development, AbbVie Deutschland GmbH & Co. KG, Knollstraße 50, 67061 Ludwigshafen, Germany
| | - Xiaojing Liu
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Hao Chen
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Xinxin Luo
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Bo Huang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Nayuan Zhou
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Hangxing Wang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Qichao Zou
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Senbiao Fang
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, China.
| | - Suxiao Wang
- Hubei Key Laboratory for Precision Synthesis of Small Molecule Pharmaceuticals; Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules; College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China.
| | - Limin Wu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| |
Collapse
|
38
|
Miao K, Xia X, Zou Y, Shi B. Small Scale, Big Impact: Nanotechnology-Enhanced Drug Delivery for Brain Diseases. Mol Pharm 2024; 21:3777-3799. [PMID: 39038108 DOI: 10.1021/acs.molpharmaceut.4c00387] [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: 07/24/2024]
Abstract
Central nervous system (CNS) diseases, ranging from brain cancers to neurodegenerative disorders like dementia and acute conditions such as strokes, have been heavily burdening healthcare and have a direct impact on patient quality of life. A significant hurdle in developing effective treatments is the presence of the blood-brain barrier (BBB), a highly selective barrier that prevents most drugs from reaching the brain. The tight junctions and adherens junctions between the endothelial cells and various receptors expressed on the cells make the BBB form a nonfenestrated and highly selective structure that is crucial for brain homeostasis but complicates drug delivery. Nanotechnology offers a novel pathway to circumvent this barrier, with nanoparticles engineered to ferry drugs across the BBB, protect drugs from degradation, and deliver medications to the designated area. After years of development, nanoparticle optimization, including sizes, shapes, surface modifications, and targeting ligands, can enable nanomaterials tailored to specific brain drug delivery settings. Moreover, smart nano drug delivery systems can respond to endogenous and exogenous stimuli that control subsequent drug release. Here, we address the importance of the BBB in brain disease treatment, summarize different delivery routes for brain drug delivery, discuss the cutting-edge nanotechnology-based strategies for brain drug delivery, and further offer valuable insights into how these innovations in nanoparticle technology could revolutionize the treatment of CNS diseases, presenting a promising avenue for noninvasive, targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kaiting Miao
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xue Xia
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Yan Zou
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bingyang Shi
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
39
|
Desai N, Chavda V, Singh TRR, Thorat ND, Vora LK. Cancer Nanovaccines: Nanomaterials and Clinical Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401631. [PMID: 38693099 DOI: 10.1002/smll.202401631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/30/2024] [Indexed: 05/03/2024]
Abstract
Cancer nanovaccines represent a promising frontier in cancer immunotherapy, utilizing nanotechnology to augment traditional vaccine efficacy. This review comprehensively examines the current state-of-the-art in cancer nanovaccine development, elucidating innovative strategies and technologies employed in their design. It explores both preclinical and clinical advancements, emphasizing key studies demonstrating their potential to elicit robust anti-tumor immune responses. The study encompasses various facets, including integrating biomaterial-based nanocarriers for antigen delivery, adjuvant selection, and the impact of nanoscale properties on vaccine performance. Detailed insights into the complex interplay between the tumor microenvironment and nanovaccine responses are provided, highlighting challenges and opportunities in optimizing therapeutic outcomes. Additionally, the study presents a thorough analysis of ongoing clinical trials, presenting a snapshot of the current clinical landscape. By curating the latest scientific findings and clinical developments, this study aims to serve as a comprehensive resource for researchers and clinicians engaged in advancing cancer immunotherapy. Integrating nanotechnology into vaccine design holds immense promise for revolutionizing cancer treatment paradigms, and this review provides a timely update on the evolving landscape of cancer nanovaccines.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502285, India
| | - Vivek Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, 380009, India
| | | | - Nanasaheb D Thorat
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
- Department of Physics, Bernal Institute, Castletroy, Limerick, V94T9PX, Ireland
- Nuffield Department of Women's & Reproductive Health, Medical Science Division, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| |
Collapse
|
40
|
Nattich-Rak M, Sadowska M, Adamczyk Z, Basinska T, Mickiewicz D, Gadzinowski M. Deposition of Human-Serum-Albumin-Functionalized Spheroidal Particles on Abiotic Surfaces: Reference Kinetic Results for Bioparticles. Molecules 2024; 29:3405. [PMID: 39064983 PMCID: PMC11279952 DOI: 10.3390/molecules29143405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Human serum albumin (HSA) corona formation on polymer microparticles of a spheroidal shape was studied using dynamic light scattering and Laser Doppler Velocimetry (LDV). Physicochemical characteristics of the albumin comprising the zeta potential and the isoelectric point were determined as a function of pH for various ionic strengths. Analogous characteristics of the polymer particles were analyzed. The adsorption of albumin on the particles was in situ monitored by LDV. The stability of the HSA-functionalized particle suspensions under various pHs and their electrokinetic properties were also determined. The deposition kinetics of the particles on mica, silica and gold sensors were investigated by optical microscopy, AFM and quartz microbalance (QCM) under diffusion and flow conditions. The obtained results were interpreted in terms of the random sequential adsorption model that allowed to estimate the range of applicability of QCM for determining the deposition kinetics of viruses and bacteria at abiotic surfaces.
Collapse
Affiliation(s)
- Małgorzata Nattich-Rak
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Cracow, Poland;
| | - Marta Sadowska
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Cracow, Poland;
| | - Zbigniew Adamczyk
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Cracow, Poland;
| | - Teresa Basinska
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Henryka Sienkiewicza 112, 90-363 Lodz, Poland; (T.B.); (D.M.); (M.G.)
| | - Damian Mickiewicz
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Henryka Sienkiewicza 112, 90-363 Lodz, Poland; (T.B.); (D.M.); (M.G.)
| | - Mariusz Gadzinowski
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Henryka Sienkiewicza 112, 90-363 Lodz, Poland; (T.B.); (D.M.); (M.G.)
| |
Collapse
|
41
|
Gao Y, Huang Y, Ren C, Chou P, Wu C, Pan X, Quan G, Huang Z. Looking back, moving forward: protein corona of lipid nanoparticles. J Mater Chem B 2024; 12:5573-5588. [PMID: 38757190 DOI: 10.1039/d4tb00186a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Lipid nanoparticles (LNPs) are commonly employed for drug delivery owing to their considerable drug-loading capacity, low toxicity, and excellent biocompatibility. Nevertheless, the formation of protein corona (PC) on their surfaces significantly influences the drug's in vivo fate (such as absorption, distribution, metabolism, and elimination) upon administration. PC denotes the phenomenon wherein one or multiple strata of proteins adhere to the external interface of nanoparticles (NPs) or microparticles within the biological milieu, encompassing ex vivo fluids (e.g., serum-containing culture media) and in vivo fluids (such as blood and tissue fluids). Hence, it is essential to claim the PC formation behaviors and mechanisms on the surface of LNPs. This overview provided a comprehensive examination of crucial aspects related to such issues, encompassing time evolution, controllability, and their subsequent impacts on LNPs. Classical studies of PC generation on the surface of LNPs were additionally integrated, and its decisive role in shaping the in vivo fate of LNPs was explored. The mechanisms underlying PC formation, including the adsorption theory and alteration theory, were introduced to delve into the formation process. Subsequently, the existing experimental outcomes were synthesized to offer insights into the research and application facets of PC, and it was concluded that the manipulation of PC held substantial promise in the realm of targeted delivery.
Collapse
Affiliation(s)
- Yue Gao
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Yeqi Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Chuanyu Ren
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Peiwen Chou
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China.
| |
Collapse
|
42
|
Qin H, Teng Y, Dai R, Wang A, Liu J. Glycan-based scaffolds and nanoparticles as drug delivery system in cancer therapy. Front Immunol 2024; 15:1395187. [PMID: 38799466 PMCID: PMC11116596 DOI: 10.3389/fimmu.2024.1395187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Glycan-based scaffolds are unique in their high specificity, versatility, low immunogenicity, and ability to mimic natural carbohydrates, making them attractive candidates for use in cancer treatment. These scaffolds are made up of glycans, which are biopolymers with well biocompatibility in the human body that can be used for drug delivery. The versatility of glycan-based scaffolds allows for the modulation of drug activity and targeted delivery to specific cells or tissues, which increases the potency of drugs and reduces side effects. Despite their promise, there are still technical challenges in the design and production of glycan-based scaffolds, as well as limitations in their therapeutic efficacy and specificity.
Collapse
Affiliation(s)
- Henan Qin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yibin Teng
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Rui Dai
- Department of Pharmacy, Peking Union Medical University Hospital, Beijing, China
| | - Aman Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
43
|
Önal Acet B, Gül D, Stauber RH, Odabaşı M, Acet Ö. A Review for Uncovering the "Protein-Nanoparticle Alliance": Implications of the Protein Corona for Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:823. [PMID: 38786780 PMCID: PMC11124003 DOI: 10.3390/nano14100823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
Understanding both the physicochemical and biological interactions of nanoparticles is mandatory for the biomedical application of nanomaterials. By binding proteins, nanoparticles acquire new surface identities in biological fluids, the protein corona. Various studies have revealed the dynamic structure and nano-bio interactions of the protein corona. The binding of proteins not only imparts new surface identities to nanoparticles in biological fluids but also significantly influences their bioactivity, stability, and targeting specificity. Interestingly, recent endeavors have been undertaken to harness the potential of the protein corona instead of evading its presence. Exploitation of this 'protein-nanoparticle alliance' has significant potential to change the field of nanomedicine. Here, we present a thorough examination of the latest research on protein corona, encompassing its formation, dynamics, recent developments, and diverse bioapplications. Furthermore, we also aim to explore the interactions at the nano-bio interface, paving the way for innovative strategies to advance the application potential of the protein corona. By addressing challenges and promises in controlling protein corona formation, this review provides insights into the evolving landscape of the 'protein-nanoparticle alliance' and highlights emerging.
Collapse
Affiliation(s)
- Burcu Önal Acet
- Faculty of Arts and Science, Chemistry Department, Aksaray University, Aksaray 68100, Turkey; (B.Ö.A.); (M.O.)
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany;
| | - Désirée Gül
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany;
| | - Roland H. Stauber
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany;
| | - Mehmet Odabaşı
- Faculty of Arts and Science, Chemistry Department, Aksaray University, Aksaray 68100, Turkey; (B.Ö.A.); (M.O.)
| | - Ömür Acet
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany;
- Vocational School of Health Science, Pharmacy Services Program, Tarsus University, Tarsus 33100, Turkey
| |
Collapse
|
44
|
Yan R, Zhan M, Xu J, Peng Q. Functional nanomaterials as photosensitizers or delivery systems for antibacterial photodynamic therapy. BIOMATERIALS ADVANCES 2024; 159:213820. [PMID: 38430723 DOI: 10.1016/j.bioadv.2024.213820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/09/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Bacterial infection is a global health problem that closely related to various diseases threatening human life. Although antibiotic therapy has been the mainstream treatment method for various bacterial infectious diseases for decades, the increasing emergence of bacterial drug resistance has brought enormous challenges to the application of antibiotics. Therefore, developing novel antibacterial strategies is of great importance. By producing reactive oxygen species (ROS) with photosensitizers (PSs) under light irradiation, antibacterial photodynamic therapy (aPDT) has emerged as a non-invasive and promising approach for treating bacterial infections without causing drug resistance. However, the insufficient therapeutic penetration, poor hydrophilicity, and poor biocompatibility of traditional PSs greatly limit the efficacy of aPDT. Recently, studies have found that nanomaterials with characteristics of favorable photocatalytic activity, surface plasmonic resonance, easy modification, and high drug loading capacity can improve the therapeutic efficacy of aPDT. In this review, we aim to provide a comprehensive understanding of the mechanism of nanomaterials-mediated aPDT and summarize the representative nanomaterials in aPDT, either as PSs or carriers for PSs. In addition, the combination of advanced nanomaterials-mediated aPDT with other therapies, including targeted therapy, gas therapy, and multidrug resistance (MDR) therapy, is reviewed. Also, the concerns and possible solutions of nanomaterials-based aPDT are discussed. Overall, this review may provide theoretical basis and inspiration for the development of nanomaterials-based aPDT.
Collapse
Affiliation(s)
- Ruijiao Yan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Meijun Zhan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jingchen Xu
- Department of Dental Medical Center, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
45
|
Ahmad S, Ahmad S, Ali S, Esa M, Khan A, Yan H. Recent Advancements and Unexplored Biomedical Applications of Green Synthesized Ag and Au Nanoparticles: A Review. Int J Nanomedicine 2024; 19:3187-3215. [PMID: 38590511 PMCID: PMC10999736 DOI: 10.2147/ijn.s453775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Green synthesis of silver (Ag) and gold (Au) nanoparticles (NPs) has acquired huge popularity owing to their potential applications in various fields. A large number of research articles exist in the literature describing the green synthesis of Ag and Au NPs for biomedical applications. However, these findings are scattered, making it time-consuming for researchers to locate promising advancements in Ag and Au NPs synthesis and their unexplored biomedical applications. Unlike other review articles, this systematic study not only highlights recent advancements in the green synthesis of Ag and Au NPs but also explores their potential unexplored biomedical applications. The article discusses the various synthesis approaches for the green synthesis of Ag and Au NPs highlighting the emerging developments and novel strategies. Then, the article reviews the important biomedical applications of green synthesized Ag and Au NPs by critically evaluating the expected advantages. To expose future research direction in the field, the article describes the unexplored biomedical applications of the NPs. Finally, the articles discuss the challenges and limitations in the green synthesis of Ag and Au NPs and their biomedical applications. This article will serve as a valuable reference for researchers, working on green synthesis of Ag and Au NPs for biomedical applications.
Collapse
Affiliation(s)
- Shahbaz Ahmad
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, People’s Republic of China
| | - Shujaat Ahmad
- Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal Dir Upper Khyber Pakhtunkhwa, Pakistan
| | - Shujat Ali
- College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Muhammad Esa
- Department of Pharmacy, Shaheed Benazir Bhutto University, Sheringal Dir Upper Khyber Pakhtunkhwa, Pakistan
| | - Ajmal Khan
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, People’s Republic of China
| | - Hai Yan
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, People’s Republic of China
| |
Collapse
|
46
|
Huang Y, Chang Z, Gao Y, Ren C, Lin Y, Zhang X, Wu C, Pan X, Huang Z. Overcoming the Low-Stability Bottleneck in the Clinical Translation of Liposomal Pressurized Metered-Dose Inhalers: A Shell Stabilization Strategy Inspired by Biomineralization. Int J Mol Sci 2024; 25:3261. [PMID: 38542235 PMCID: PMC10970625 DOI: 10.3390/ijms25063261] [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: 02/21/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 06/25/2024] Open
Abstract
Currently, several types of inhalable liposomes have been developed. Among them, liposomal pressurized metered-dose inhalers (pMDIs) have gained much attention due to their cost-effectiveness, patient compliance, and accurate dosages. However, the clinical application of liposomal pMDIs has been hindered by the low stability, i.e., the tendency of the aggregation of the liposome lipid bilayer in hydrophobic propellant medium and brittleness under high mechanical forces. Biomineralization is an evolutionary mechanism that organisms use to resist harsh external environments in nature, providing mechanical support and protection effects. Inspired by such a concept, this paper proposes a shell stabilization strategy (SSS) to solve the problem of the low stability of liposomal pMDIs. Depending on the shell material used, the SSS can be classified into biomineralization (biomineralized using calcium, silicon, manganese, titanium, gadolinium, etc.) biomineralization-like (composite with protein), and layer-by-layer (LbL) assembly (multiple shells structured with diverse materials). This work evaluated the potential of this strategy by reviewing studies on the formation of shells deposited on liposomes or similar structures. It also covered useful synthesis strategies and active molecules/functional groups for modification. We aimed to put forward new insights to promote the stability of liposomal pMDIs and shed some light on the clinical translation of relevant products.
Collapse
Affiliation(s)
- Yeqi Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (Y.H.); (Y.G.); (C.R.); (Y.L.); (C.W.)
| | - Ziyao Chang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (X.P.)
| | - Yue Gao
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (Y.H.); (Y.G.); (C.R.); (Y.L.); (C.W.)
| | - Chuanyu Ren
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (Y.H.); (Y.G.); (C.R.); (Y.L.); (C.W.)
| | - Yuxin Lin
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (Y.H.); (Y.G.); (C.R.); (Y.L.); (C.W.)
| | - Xuejuan Zhang
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (Y.H.); (Y.G.); (C.R.); (Y.L.); (C.W.)
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (Y.H.); (Y.G.); (C.R.); (Y.L.); (C.W.)
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (X.P.)
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (Y.H.); (Y.G.); (C.R.); (Y.L.); (C.W.)
| |
Collapse
|
47
|
Zhuo X, Wu Y, Fu X, Li J, Xiang Y, Liang X, Mao C, Jiang Y. Genome editing of PAR2 through targeted delivery of CRISPR-Cas9 system for alleviating acute lung inflammation via ERK/NLRP3/IL-1 β and NO/iNOS signalling. Acta Pharm Sin B 2024; 14:1441-1456. [PMID: 38487002 PMCID: PMC10935474 DOI: 10.1016/j.apsb.2023.08.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 03/17/2024] Open
Abstract
Excessive and uncontrollable inflammatory responses in alveoli can dramatically exacerbate pulmonary disease progressions through vigorous cytokine releases, immune cell infiltration and protease-driven tissue damages. It is an urgent need to explore potential drug strategies for mitigating lung inflammation. Protease-activated receptor 2 (PAR2) as a vital molecular target principally participates in various inflammatory diseases via intracellular signal transduction. However, it has been rarely reported about the role of PAR2 in lung inflammation. This study applied CRISPR-Cas9 system encoding Cas9 and sgRNA (pCas9-PAR2) for PAR2 knockout and fabricated an anionic human serum albumin-based nanoparticles to deliver pCas9-PAR2 with superior inflammation-targeting efficiency and stability (TAP/pCas9-PAR2). TAP/pCas9-PAR2 robustly facilitated pCas9-PAR2 to enter and transfect inflammatory cells, eliciting precise gene editing of PAR2 in vitro and in vivo. Importantly, PAR2 deficiency by TAP/pCas9-PAR2 effectively and safely promoted macrophage polarization, suppressed pro-inflammatory cytokine releases and alleviated acute lung inflammation, uncovering a novel value of PAR2. It also revealed that PAR2-mediated pulmonary inflammation prevented by TAP/pCas9-PAR2 was mainly dependent on ERK-mediated NLRP3/IL-1β and NO/iNOS signalling. Therefore, this work indicated PAR2 as a novel target for lung inflammation and provided a potential nanodrug strategy for PAR2 deficiency in treating inflammatory diseases.
Collapse
Affiliation(s)
- Xin Zhuo
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yue Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiujuan Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jianbin Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yuxin Xiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaoyu Liang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Canquan Mao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
48
|
Veider F, Sanchez Armengol E, Bernkop-Schnürch A. Charge-Reversible Nanoparticles: Advanced Delivery Systems for Therapy and Diagnosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304713. [PMID: 37675812 DOI: 10.1002/smll.202304713] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/24/2023] [Indexed: 09/08/2023]
Abstract
The past two decades have witnessed a rapid progress in the development of surface charge-reversible nanoparticles (NPs) for drug delivery and diagnosis. These NPs are able to elegantly address the polycation dilemma. Converting their surface charge from negative/neutral to positive at the target site, they can substantially improve delivery of drugs and diagnostic agents. By specific stimuli like a shift in pH and redox potential, enzymes, or exogenous stimuli such as light or heat, charge reversal of NP surface can be achieved at the target site. The activated positive surface charge enhances the adhesion of NPs to target cells and facilitates cellular uptake, endosomal escape, and mitochondrial targeting. Because of these properties, the efficacy of incorporated drugs as well as the sensitivity of diagnostic agents can be essentially enhanced. Furthermore, charge-reversible NPs are shown to overcome the biofilm formed by pathogenic bacteria and to shuttle antibiotics directly to the cell membrane of these microorganisms. In this review, the up-to-date design of charge-reversible NPs and their emerging applications in drug delivery and diagnosis are highlighted.
Collapse
Affiliation(s)
- Florina Veider
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| | - Eva Sanchez Armengol
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| | - Andreas Bernkop-Schnürch
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| |
Collapse
|
49
|
Chakraborty P, Bhattacharyya C, Sahu R, Dua TK, Kandimalla R, Dewanjee S. Polymeric nanotherapeutics: An emerging therapeutic approach for the management of neurodegenerative disorders. J Drug Deliv Sci Technol 2024; 91:105267. [DOI: 10.1016/j.jddst.2023.105267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
50
|
Chen G, Yang Z, Yu X, Yu C, Sui S, Zhang C, Bao C, Zeng X, Chen Q, Peng Q. Intratumor delivery of amino-modified graphene oxide as a multifunctional photothermal agent for efficient antitumor phototherapy. J Colloid Interface Sci 2023; 652:1108-1116. [PMID: 37657211 DOI: 10.1016/j.jcis.2023.08.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 09/03/2023]
Abstract
Due to the high selectivity and non-invasive property, phototherapy has attracted increasing attention in the treatment of cancer. Targeted delivery and retention of photoactive agents in tumor tissue is of great significance and importance for safe and efficient phototherapy. Herein, we report a multifunctional nanomaterial photothermal agent, namely amino-modified graphene oxide (AGO) for anti-oral cancer photothermal therapy (PTT). Compared to the parental graphene oxide (GO) which has a negative charge and weak photothermal effect, AGO possesses a positive charge (∼+50 mV) and the significantly enhanced photothermal effect. Positive charge allows AGO to efficiently interact with tumor cells and retain in tumor tissue after intratumor injection. The enhanced photothermal effect allows AGO to achieve the tunable and efficient PTT. In vitro results show that AGO (15 μg/mL) reduces the viability of HSC-3 cells (oral squamous cell carcinoma cell line) to 5% under near infrared (NIR) irradiation (temperature increased to 58.4 °C). In vivo antitumor study shows that intratumor delivery of AGO (200 μg/mouse) has no inhibition effects on tumor growth (454% of initial tumor size) without NIR. With a single dose of NIR irradiation, however, AGO significantly reduces the tumor size to 25% of initial size in 1 of 4 mice, and even induces the complete tumor ablation in 3 of 4 mice. Furthermore, the injected AGO falls off along with the scab after PTT. Our findings indicate that AGO is a potential nano-photothermal agent for tunable, convenient and efficient anticancer PTT.
Collapse
Affiliation(s)
- Geyun Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhenghao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiaotong Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chenhao Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shangyan Sui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chaoliang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|