1
|
Ghosh D, Guin A, Kumar A, Das A, Paul S. Comprehensive insights of etiological drivers of hepatocellular carcinoma: Fostering targeted nano delivery to anti-cancer regimes. Biochim Biophys Acta Rev Cancer 2025; 1880:189318. [PMID: 40222420 DOI: 10.1016/j.bbcan.2025.189318] [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: 10/30/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Hepatocellular carcinoma (HCC) stands as one of the most prevalent and deadliest malignancies on a global scale. Its complex pathogenesis arises from multifactorial etiologies, including viral infections, metabolic syndromes, and environmental carcinogens, all of which drive genetic and molecular aberrations in hepatocytes. This intricate condition is associated with multiple causative factors, resulting in the abnormal activation of various cellular and molecular pathways. Given that HCC frequently manifests within the context of a compromised or cirrhotic liver, coupled with the tendency of late-stage diagnoses, the overall prognosis tends to be unfavorable. Systemic therapy, especially conventional cytotoxic drugs, generally proves ineffective. Despite advancements in therapeutic interventions, conventional treatments such as chemotherapy often exhibit limited efficacy and substantial systemic toxicity. In this context, nanomedicine, particularly lipid-based nanoparticles (LNPs), has emerged as a promising strategy for enhancing drug delivery specificity and reducing adverse effects. This review provides a comprehensive overview of the molecular and metabolic underpinnings of HCC. Furthermore, we explored the role of lipid-based nano-formulations including liposomes, solid lipid nanoparticles, and nanostructured lipid carriers in targeted drug delivery for HCC. We have highlighted recent advances in LNP-based delivery approaches, FDA-approved drugs, and surface modification strategies to improve liver-specific delivery and therapeutic efficacy. It will provide a comprehensive summary of various treatment strategies, recent clinical advances, receptor-targeting strategies and the role of lipid composition in cellular uptake. The review concludes with a critical assessment of existing challenges and future prospects in nanomedicines-driven HCC therapy.
Collapse
Affiliation(s)
- Dipanjan Ghosh
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Aharna Guin
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Aryan Kumar
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Amlan Das
- Department of Microbiology & Department of Biochemistry, Royal School of Biosciences, The Assam Royal Global University, Guwahati 781035, Assam, India.
| | - Santanu Paul
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India.
| |
Collapse
|
2
|
Gil-Vives M, Hernández M, Hernáez Á, Borrós S, Fornaguera C. Safety of nanoparticle therapies during pregnancy: A systematic review and meta-analysis. J Control Release 2025; 382:113655. [PMID: 40122240 DOI: 10.1016/j.jconrel.2025.113655] [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: 01/22/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
The exclusion of pregnant women from clinical trials has led to insufficient safety data for many treatments, making it necessary to evaluate their potential benefits and risks during preclinical stages. Nanomedicines show potential for reduced toxicity but there is limited evidence about their safety for pregnant women and their fetuses. We conducted the first systematic review and meta-analysis of the effect of nanoparticles (NPs) on a key outcome of fetal toxicity (low birth weight) in murine models. In the meta-analysis of mouse models, negatively charged NPs tended to decrease birth weight (-69.8 mg, 95 % CI: -196 to 56.5), as did small (-191 mg, 95 % CI: -369 to -13.3) and plain inorganic nanosystems (-249 mg, 95 % CI: -535 to 37.4). In contrast, positively charged NPs resulted in increased birth weight (+29.3 mg, 95 % CI: 23.4 to 35.2). All findings were validated in studies with low heterogeneity and low risk of publication bias. Neither large NPs (+4.37 mg; 95 % CI: -45.3 to 54.0) nor polymer-coated NPs (+16.5 mg; 95 % CI: -44.7 to 77.6) had any clear association with birth weight. Similar results were observed in other models and experimental designs from articles not included in the meta-analysis, although no conclusions were drawn for other parameters due to high variability. Our findings pave the way for future research and the rational development of safer nanomedicines for use during pregnancy.
Collapse
Affiliation(s)
- Maria Gil-Vives
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Via Augusta 390, 08017 Barcelona, Spain; Blanquerna School of Health Sciences, Universitat Ramon Llull (URL), Padilla 326, 08025 Barcelona, Spain
| | - Marta Hernández
- Blanquerna School of Health Sciences, Universitat Ramon Llull (URL), Padilla 326, 08025 Barcelona, Spain.
| | - Álvaro Hernáez
- Blanquerna School of Health Sciences, Universitat Ramon Llull (URL), Padilla 326, 08025 Barcelona, Spain; Hospital del Mar Research Institute (IMIM), Doctor Aiguader 88, 08003 Barcelona, Spain.; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Av. Monforte de Lemos 5, 28029 Madrid, Spain
| | - Salvador Borrós
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Via Augusta 390, 08017 Barcelona, Spain
| | - Cristina Fornaguera
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Via Augusta 390, 08017 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys 23, 08010 Barcelona, Spain..
| |
Collapse
|
3
|
Ruan Y, Liu Y, Lei G, Shao Y, Zhao Y, Gong J, Li K, Xie X, Wang S, Sun X. Fluoroalkane Engineered Magnetic Vectors Unlock the Potential of Gasdermin in Vivo Delivery for Pyroptosis Induced Cancer Therapy. Adv Healthc Mater 2025; 14:e2500801. [PMID: 40231596 DOI: 10.1002/adhm.202500801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/19/2025] [Indexed: 04/16/2025]
Abstract
Pyroptosis, a programmed necrotic cell death mediated by gasdermin, can activate strong immune responses and serve as a potential target for cancer therapy. Nevertheless, the relatively large molecular size and negative surface charge of gasdermin impede them from effectively intracellular delivery and directly inducing pyroptosis. Here, a cytosolic protein delivery system, fluorinated iron oxide nanoparticles (FIONPs) is reported, which can self-assemble with active gasdermin A3 protein (GSDMA3) via noncovalent interactions and effectively trigger pyroptosis in 4T1 cells. It is proved that the delivery system is versatile for various cargo proteins (ribonuclease A, saporin, β-galactosidase, and bovine serum albumin) with different isoelectric points and molecular weights, without compromising their biological activity in vitro. What's more, under magnetic drive, FIONPs facilitate active transport of GSDMA3 in vivo, further augmenting tumor suppression and immune response. Overall, magnetic-driven FIONPs provide an effective delivery system for intracellular protein transductions, and the application of the delivery system reveals that direct delivery of GSDMA3 significantly elicits robust antitumor immunity via the induction of pyroptosis.
Collapse
Affiliation(s)
- Yiling Ruan
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Guojun Lei
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yuan Shao
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yaxuan Zhao
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Jinglang Gong
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Keying Li
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Xiaojiang Xie
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, P. R. China
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, P. R. China
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, P. R. China
| |
Collapse
|
4
|
Choi S, Choi H, Chung JW, Kim SH. Injectable Endoplasmin-Loaded Lipid Nanoparticles-Hydrogel Composite for Cartilage Regeneration. Tissue Eng Regen Med 2025; 22:409-424. [PMID: 39992620 PMCID: PMC12122951 DOI: 10.1007/s13770-024-00698-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/12/2024] [Accepted: 12/22/2024] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Endoplasmin (ENPL), a heat shock protein 90 family member, promotes chondrogenic differentiation of stem cells by inhibiting ERK1/2 phosphorylation and inducing endoplasmic reticulum stress. However, its large size limits cellular uptake and therapeutic potential. To overcome this challenge, a cationic lipid nanoparticle (C_LNP) system was designed to deliver ENPL intracellularly, enhancing its effects on human tonsil-derived mesenchymal stem cells (hTMSCs). METHODS ENPL-loaded cationic lipid nanoparticles (ENPL_C_LNP) were synthesized to facilitate intracellular ENPL delivery. The delivery efficiency and cytotoxicity were assessed in vitro using hTMSCs. Additionally, ENPL_C_LNPs were incorporated into a hyaluronic acid and chondroitin sulfate-based injectable hydrogel and tested for chondrogenic differentiation potential in a mouse subcutaneous model. RESULTS ENPL_C_LNP achieved over 80% intracellular protein delivery efficiency with no cytotoxic effects. Co-cultured hTMSCs exhibited increased glycosaminoglycans (GAGs) and collagen expression over 21 days. In vivo, the hydrogel-embedded ENPL_C_LNP system enabled stable cartilage differentiation, evidenced by abundant cartilage-specific lacuna structures in regenerated tissue. CONCLUSION Combining ENPL_C_LNP with an injectable hydrogel scaffold supports chondrogenic differentiation and cartilage regeneration, offering a promising strategy for cartilage tissue engineering.
Collapse
Affiliation(s)
- Sumi Choi
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan, 49315, Republic of Korea
| | - Hyeongrok Choi
- Department of Biomedical Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Jin Woong Chung
- Department of Biomedical Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
5
|
Ranjan A, Rajput VD, Shende S, Saxena P, Prazdnova EV, Sushkova S, Arora J, Chauhan A, Jindal T, Zargar SM, Minkina T. Eco-friendly fabrication of Zn-based nanoparticles: implications in agricultural advancement and elucidation of toxicity aspects. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:4130-4167. [PMID: 40189734 DOI: 10.1002/jsfa.14243] [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: 08/29/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 05/17/2025]
Abstract
Zinc (Zn) is a vital micronutrient required for optimal plant growth and soil fertility. Its use in the form of nanoparticles (NPs) has gained significant attention in agricultural applications. Green synthesized Zn-based NPs offer an eco-friendly solution to several conventional problems in agriculture. Several plants, bacteria, fungi and yeast have shown significant potential in fabricating Zn NPs that can provide environmentally friendly solutions in agriculture and the approach is aligned with sustainable agricultural practices, reducing the dependency on harmful agrochemicals. Zn-based NPs act as plant growth promoters, enhance crop yield, promote resilience to abiotic stressors and are efficient crop protection agents. Their role as a smart delivery system, enabling targeted and controlled release of agrochemicals, further signifies their potential use in agriculture. Because agriculture requires repeated applications hence, the toxicological aspects of Zn NPs cannot be ignored. Zn NPs are reported to cause phytotoxicity, including root damage, physiological and biochemical disturbances, and genotoxic effects. Furthermore, exposure to Zn NPs poses risks to soil microbiota, and aquatic and terrestrial organisms potentially impacting the ecosystem. The green synthesis of Zn-based NPs has a promising aspect for advancing sustainable agriculture by reducing agrochemical use and improving crop productivity. Their diverse applications as plant growth promoters, crop protectants and smart delivery systems emphasize their potential. However, the toxicological aspects are essential to ensure the standardization of doses for their safe and effective use. Further research would help address such concerns and help in developing viable and eco-friendly solutions for modern agriculture. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, India
| | - Vishnu D Rajput
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Sudhir Shende
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
- Department of Microbiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland
| | - Pallavi Saxena
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
- Adjunct Faculty, Centre for Research and Outcome, Chitkara University, Rajpura, India
| | - Evgeniya V Prazdnova
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Svetlana Sushkova
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Jayati Arora
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, India
| | - Tanu Jindal
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, India
| | - Sajad Majeed Zargar
- Proteomics Lab, Division of Plant Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, India
| | - Tatiana Minkina
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| |
Collapse
|
6
|
Masoumi S, Aleyasin SA, Faghihi S. Albumin nanoparticles-mediated doxorubicin delivery enhances the anti-tumor efficiency in ovarian cancer cells through controlled release. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6885-6900. [PMID: 39692901 DOI: 10.1007/s00210-024-03730-3] [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: 06/28/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
Doxorubicin (DOX) is an anthracycline commonly used as a first-line treatment option for various malignancies, either as a stand-alone treatment or in combination with other chemotherapeutic agents. However, its efficacy in advanced cancer stages requires high doses, resulting in significant cytotoxicity to normal cells and severe side effects. Nanotechnology offers a promising strategy to mitigate these drawbacks through controlled drug release. In this study, bovine serum albumin nanoparticles (BSA-NPs) were synthesized via the desolvation method and successfully loaded with DOX (DOX-BSA-NPs). Characterization using dynamic light scattering, scanning electron microscopy, Fourier-transform infrared spectroscopy, UV-visible spectroscopy, and high-performance liquid chromatography confirmed efficient drug loading. In vitro studies demonstrated that DOX-BSA-NPs enabled sustained drug release and enhanced intracellular delivery. After treatment with DOX-BSA-NPs, ovarian cancer cells showed a twofold increase in cytotoxicity compared to free DOX. Scratch assays further revealed a significant reduction in cancer cell migration and invasion. Additionally, LDH assays and Annexin V-FITC flow cytometry indicated a shift toward apoptosis over necrosis, enhancing the anti-tumor efficacy of DOX. This was supported by increased reactive oxygen species production, upregulation of pro-apoptotic genes, downregulation of anti-apoptotic genes, and elevated caspase 3 and 7 activity, collectively promoting apoptosis. These findings underscore the potential of DOX-BSA-NPs as a superior alternative for targeted and controlled drug delivery, offering enhanced therapeutic efficacy and reduced side effects in ovarian cancer treatment.
Collapse
Affiliation(s)
- Sajjad Masoumi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Seyed Ahmad Aleyasin
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| | - Shahab Faghihi
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
7
|
Zeng H, Li D, He Q, Zheng X, Chen X, Jian G, Zhang H, Chen T. A microenvironment responsive nanoparticle regulating osteoclast fate to promote bone repair in osteomyelitis. Mater Today Bio 2025; 32:101777. [PMID: 40321696 PMCID: PMC12049845 DOI: 10.1016/j.mtbio.2025.101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/30/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Osteomyelitis exhibits bone defects in an inflammatory and acid microenvironment. As a crucial factor in this inflammation responses, the macrophage-osteoclast axis is absolutely the core to regulate. The research explored a shell-core structured biomaterial, consisting of a gelatin nanoparticle (GNP) platform loaded with bone morphogenetic protein 9 (BMP9) and coated with a metal phenolic network (TA-Ce), which exhibited adaptive sensitivity to pH values. Extracellularly, it rapidly responded to lower pH, achieving specific release in an inflammatory microenvironment. Intracellularly, it impacted the formation, function, and differentiation of osteoclasts through the macrophage-osteoclast axis, thereby promoting bone defect repair. In vivo and in vitro studies showed GNPs-BMP9@TA-Ce regulated osteoclasts to optimize osteomyelitis treatment strategies, highlighting the potential of modified nanobiomaterials for clinical application.
Collapse
Affiliation(s)
- Huan Zeng
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
- Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Dize Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Qingqing He
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xinhui Zheng
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xu Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Guangyu Jian
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Hongmei Zhang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
- Department of Pediatric Dentistry, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Tao Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| |
Collapse
|
8
|
Dietz L, Simon J, Speth KR, Landfester K, Mailänder V. Plasma protein corona on silica nanoparticles enhances exocytosis. Biomater Sci 2025. [PMID: 40433828 DOI: 10.1039/d4bm01189a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
While the influence of the protein corona on nanoparticle uptake in mammalian cells is well understood, little is known about the influence of the protein corona on nanoparticle exocytosis. However, the exocytosis of nanoparticles also contributes to the therapeutic efficacy as it influences the net delivery of nanoparticles to a cell. In this study we demonstrate that the exocytosis of silica nanoparticles from HCT 116 cells is enhanced by the pre-adsorption of a human plasma protein corona. This pre-adsorption effect also depends on the diameter of the nanoparticles. The exocytosis of small silica nanoparticles (10 nm) is less pronounced, while the exocytosis of larger silica nanoparticles (100 nm) is significantly increased in the presence of a protein corona. A proteomic analysis of the plasma protein corona of the different-sized silica nanoparticles (10 nm, 30 nm, 50 nm, and 100 nm) reveals different protein compositions. Apolipoproteins and coagulation proteins are enriched in a size-dependent manner with high amounts of apolipoproteins adsorbed to small silica nanoparticles. The findings underscore the significance of the nanoparticle protein corona for exocytosis and demonstrate the need to engineer nanocarriers that are not exocytosed rapidly to enhance the efficacy in drug delivery.
Collapse
Affiliation(s)
- Laura Dietz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Julia Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Kai R Speth
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
9
|
Chen Q, Yu Y, Tong L, Weiss RM, Wei SG. Targeted delivery of TAPI-1 via biomimetic nanoparticles ameliorates post-infarct left ventricle function and remodelling. Cardiovasc Res 2025; 121:760-774. [PMID: 40038918 PMCID: PMC12101320 DOI: 10.1093/cvr/cvaf039] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/21/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
AIMS The potential of nanoparticles as effective drug delivery tools for treating failing hearts in heart failure remains a challenge. Leveraging the rapid infiltration of neutrophils into infarcted hearts after myocardial infarction (MI), we developed a nanoparticle platform engineered with neutrophil membrane proteins for the targeted delivery of TAPI-1, a TACE/ADAM17 inhibitor, to the inflamed myocardium, aiming to treat cardiac dysfunction and remodelling in rats with MI. METHODS AND RESULTS Neutrophil-mimic liposomal nanoparticles (Neu-LNPs) were constructed by integrating synthesized liposomal nanoparticles with LPS-stimulated neutrophil membrane fragments and then loaded with TAPI-1. MI rats were treated with TAPI-1 delivered via Neu-LNPs for 4 weeks. Left ventricular function was assessed by echocardiography and cardiac fibrosis was evaluated post-treatment. The novel Neu-LNPs maintained typical nanoparticle features, but with increased biocompatibility. Neu-LNPs demonstrated improved targeting ability and cellular internalization, facilitated by LFA1/Mac1/ICAM-1 interaction. Neu-LNPs displayed higher accumulation and cellular uptake by macrophages and cardiomyocytes in infarcted hearts post-MI, with a sustained duration. Treatments with TAPI-1-Neu-LNPs demonstrated greater protection against myocardial injury and cardiac dysfunction in MI rats compared to untargeted TAPI-1, along with reduced cardiac collagen deposition and expression of fibrosis biomarkers as well as altered immune cell compositions within the hearts. CONCLUSIONS Targeted treatment with TACE/ADAM17 inhibitor delivered via biomimetic nanoparticles exhibited pronounced advantages in improving left ventricle function, mitigating cardiac remodelling, and reducing inflammatory responses within the infarcted hearts. This study underscores the effectiveness of Neu-LNPs as a drug delivery strategy to enhance therapeutic efficacy in clinical settings.
Collapse
Affiliation(s)
- Qing Chen
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Yang Yu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Lei Tong
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, 501 Newton Road, Iowa City, IA 52242, USA
- Veteran Affairs Medical Center, Research and Development, 601 HWY 6 WEST, Iowa City, IA 52246, USA
| | - Shun-Guang Wei
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, 501 Newton Road, Iowa City, IA 52242, USA
- Veteran Affairs Medical Center, Research and Development, 601 HWY 6 WEST, Iowa City, IA 52246, USA
- Iowa Neuroscience Institute, University of Iowa, 169 Newton Road, Iowa City, IA 52242, USA
| |
Collapse
|
10
|
Noguchi H. Curvature-sensing and generation by membrane proteins: a review. SOFT MATTER 2025; 21:3922-3940. [PMID: 40302616 DOI: 10.1039/d5sm00101c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Membrane proteins are crucial in regulating biomembrane shapes and controlling the dynamic changes in membrane morphology during essential cellular processes. These proteins can localize to regions with their preferred curvatures (curvature sensing) and induce localized membrane curvature. Thus, this review describes the recent theoretical development in membrane remodeling performed by membrane proteins. The mean-field theories of protein binding and the resulting membrane deformations are reviewed. The effects of hydrophobic insertions on the area-difference elasticity energy and that of intrinsically disordered protein domains on the membrane bending energy are discussed. For the crescent-shaped proteins, such as Bin/Amphiphysin/Rvs superfamily proteins, anisotropic protein bending energy and orientation-dependent excluded volume significantly contribute to curvature sensing and generation. Moreover, simulation studies of membrane deformations caused by protein binding are reviewed, including domain formation, budding, and tubulation.
Collapse
Affiliation(s)
- Hiroshi Noguchi
- Institute for Solid State Physics, University of Tokyo, Kashiwa, Chiba 277-8581, Japan.
| |
Collapse
|
11
|
Sahli C, Kenry. The Journey and Modes of Action of Therapeutic Nanomaterials in Cells. Bioconjug Chem 2025; 36:914-929. [PMID: 40213918 DOI: 10.1021/acs.bioconjchem.4c00584] [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: 05/22/2025]
Abstract
Over past decades, a wide range of nanomaterials have been synthesized and exploited to augment the efficacy and biocompatibility of disease theranostics and nanomedicine. The unique physicochemical properties of nanomaterials, such as high specific surface area, tunable size and shape, and versatile surface chemistry, enable the controlled modulation of nanomaterial-biosystem interactions and, consequently, more precise interventions, particularly at the cellular level. The selective modulation of nanomaterial-cell interactions can be leveraged to regulate cellular internalization, intracellular trafficking and localization, and cellular clearance of nanomaterials to enhance the disease therapeutic efficacy and minimize potential cytotoxicity. Herein, we provide an overview of our recent understanding of the journey and modes of action of therapeutic nanomaterials in cells. Specifically, we highlight the various pathways of cellular internalization, trafficking, and excretion of these nanomaterials. The different modes of action of therapeutic nanomaterials, especially controlled release and delivery, photothermal and photodynamic effects, and immunomodulation, are also discussed. We conclude our review by offering some perspectives on the current challenges and potential opportunities in this field.
Collapse
Affiliation(s)
- Célia Sahli
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Kenry
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
- Clinical and Translational Oncology Program and Skin Cancer Institute, University of Arizona Cancer Center, University of Arizona, Tucson, Arizona 85721, United States
- BIO5 Institute, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
12
|
Ngo TH, Menon S, Rivero-Müller A. Nano-immunotherapy: Merging immunotherapy precision with nanomaterial delivery. iScience 2025; 28:112319. [PMID: 40292310 PMCID: PMC12033950 DOI: 10.1016/j.isci.2025.112319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
In current landscape of cancer treatment, nanotherapy and cellular therapy stand out as promising and innovative approaches. Nanotherapy have excelled in delivering functional molecules effectively to target cancer cells, however the targetability is mostly the result of the enhanced permeability and retention effect. Meanwhile, cellular therapies such recently emerging chimeric antigen receptor (CAR)-T therapy are proficient at specifically targeting cancer cells by using engineered receptors on T cells. Yet, cellular therapies preform poor in solid tumors due to immunosuppression and cancer cell resistance to immuno-stimulation, in other words their delivery of deadly cargo is deficient. Therefore, combining nanotherapy and immunotherapy is an emerging trend, with ongoing clinical trials exploring their synergistic effects. This 2-input approach holds promise for enhancing treatment efficacy and overcoming limitations in cancer therapy. In this review, we will discuss two aspects: targetability and delivery for each individual therapy and what the combined nano-immunotherapy strategies have achieved up to now. In the last section, some future perspectives for this combination are suggested.
Collapse
Affiliation(s)
- Thu Ha Ngo
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Soumya Menon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
13
|
Męczyńska-Wielgosz S, Sikorska K, Czerwińska M, Kapka-Skrzypczak L, Kruszewski M. Uptake and Toxicity of Polystyrene NPs in Three Human Cell Lines. Int J Mol Sci 2025; 26:4783. [PMID: 40429925 PMCID: PMC12111990 DOI: 10.3390/ijms26104783] [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: 04/17/2025] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Internalization of nanoparticles (NPs), including nanoplastic, is one of the key factors determining their toxicity. In this work, we studied the toxicity and mechanisms of the uptake of model fluorescent polystyrene NPs (PS NPs) of three different sizes (30, 50, and 100 nm) in three human cancer cells lines; two originated from gut tissue (HT-29 and Caco-2) and one originated from liver tissue (Hep G2). Toxicity was measured by Neutral Red Assay (NRU), whereas mechanisms of uptake were studied using flow cytometry and different uptake inhibitors. The toxicity of the studied NPs followed a general rule observed for NPs-the smaller ones were more toxic than the larger ones. This relationship was dose dependent; however, the overall toxicity of the studied NPs was very low, despite the significant uptake of PS NPs. Although clathrin- and caveolin-dependent uptake is generally accepted as a major route of NP uptake, the inhibition of both mechanisms did not affect PS NP uptake in the cell lines studied in this work. Further experiments revealed that the major route of PS NP uptake in these cells is a scavenger receptor-mediated uptake.
Collapse
Affiliation(s)
- Sylwia Męczyńska-Wielgosz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (S.M.-W.); (K.S.); (M.C.)
| | - Katarzyna Sikorska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (S.M.-W.); (K.S.); (M.C.)
| | - Malwina Czerwińska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (S.M.-W.); (K.S.); (M.C.)
| | - Lucyna Kapka-Skrzypczak
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090 Lublin, Poland;
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (S.M.-W.); (K.S.); (M.C.)
- Department of Molecular Biology and Translational Research, Institute of Rural Health, 20-090 Lublin, Poland;
| |
Collapse
|
14
|
Zhang Y, Du Y, Zhou S, Liu Z, Li P, Du Z. Topical application of insulin encapsulated by chitosan-modified PLGA nanoparticles to alleviate alkali burn-induced corneal neovascularization. NANOSCALE 2025; 17:12323-12339. [PMID: 40278870 DOI: 10.1039/d4nr05507a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Corneal neovascularization (CRNV) severely impairs corneal transparency and is one of the leading causes of vision loss worldwide. Drug therapy is the main approach to inhibit CRNV. Insulin (INS) has been reported to facilitate the healing of corneal injuries and suppress inflammation. However, but due to the unique physiological barriers of the eye, its bioavailability is low, limiting its therapeutic effect. In this study, we developed a chitosan-poly(lactic-co-glycolic acid)-INS nanoparticles (CPI NPs) system for INS delivery. The characterization of CPI NPs was satisfactory. Experimental results demonstrated that CPI NPs effectively inhibited the migration of vascular endothelial cells and the formation of tubular structures. Furthermore, CPI NPs markedly suppressed the neovascularization in a CRNV model without any observable side effects. Quantitative proteomics analysis indicated that INS treatment led to a reduction in FTO levels within the neovascularized cornea. Both in vitro and in vivo experiments substantiated the impact of CPI NPs on FTO protein expression and the N6-methyladenosine modification. In conclusion, this study successfully developed an effective ocular drug delivery system for the treatment of CRNV induced by alkali burns, thereby offering a novel therapeutic option for this condition.
Collapse
Affiliation(s)
- Yuqing Zhang
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, China
| | - Yangrui Du
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Sijie Zhou
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Zeqi Liu
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhiyu Du
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
15
|
Bussin B, MacDuff MGG, Ngo W, Wu JLY, Lin ZP, Granda Farias A, Stordy B, Sepahi Z, Ahmed S, Moffat J, Chan WCW. Discovering nanoparticle corona ligands for liver macrophage capture. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01903-6. [PMID: 40374797 DOI: 10.1038/s41565-025-01903-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/12/2025] [Indexed: 05/18/2025]
Abstract
Liver macrophages capture circulating nanoparticles and reduce their delivery to target organs. Serum proteins adsorb to the nanoparticle surface after administration. However, the adsorbed serum proteins and their cognate cell receptors for removing nanoparticles from the bloodstream have not been linked. Here we use a multi-omics strategy to identify the adsorbed serum proteins binding to specific liver macrophage receptors. We discovered six absorbed serum proteins that bind to two liver macrophage receptors. Nanoparticle physicochemical properties can affect the degree of the six serum proteins adsorbing to the surface, the probability of binding to cell receptors and whether the liver removes the nanoparticle from circulation. Identifying the six adsorbed proteins allowed us to engineer decoy nanoparticles that prime the liver to take up fewer therapeutic nanoparticles, enabling more nanoparticles for targeting extrahepatic tissues. Elucidating the molecular interactions governing the nanoparticle journey in vivo will enable us to control nanoparticle delivery to diseased tissues.
Collapse
Affiliation(s)
- Bram Bussin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Marshall G G MacDuff
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Wayne Ngo
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Gladstone Institutes, San Francisco, CA, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Jamie L Y Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Zachary P Lin
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Granda Farias
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Stordy
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Zahra Sepahi
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Sara Ahmed
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- MD/PhD Program, University of Toronto, Toronto, Ontario, Canada
| | - Jason Moffat
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
16
|
Kaushal K, Kapoor DU, Kumar S, Sony A, Viswanath A, Chaitanya MVNL, Singh M, Singh SK, Mazumder A. Natural sesquiterpene lactones in prostate cancer therapy: mechanisms and sources. Med Oncol 2025; 42:212. [PMID: 40372575 DOI: 10.1007/s12032-025-02740-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025]
Abstract
Prostate cancer is a condition characterized by the uncontrolled proliferation of abnormal cells inside the prostate gland, part of the male reproductive system. Prostate cancer is the most common cancer among men and the second largest cause of cancer-related mortality in the United States. A novel approach to treating advanced Prostate cancer has emerged, attributable to the enhanced effectiveness of new pharmacological agents sourced from natural origins and this has led to increased rates of global existence and progression-free survival. Sesquiterpene lactones and their derivatives are now used worldwide to create and manufacture innovative cancer therapeutics. A thorough search was performed according to PRISMA guidelines in SciMed, PubMed, and Google Scholar, focusing on publications published from 1999 to 2024. The safety, efficacy, and bioactivity of sesquiterpene lactones must be evaluated via clinical trials, in vitro studies, and in vivo research and data was rigorously gathered and validated to verify its accuracy and usefulness. Prostate cancer may be treated far more effectively using naturally occurring sesquiterpene lactone molecules. The most prominent sesquiterpene lactones identified were artemisinin, alantolactone, costunolide, helenalin, cynaropicrin, parthenolide, and inuviscolide, which are originated from botanical sources like Ferula penninervis, Tanacetum argenteum, Artemisia kopetdaghensis, Cichorium intybus, Carpesium divaricatum, and Leptocarpha rivularis. Numerous studies indicated that sesquiterpene lactones may treat cancer by modifying many cellular signaling pathways, including PI3K/AKT, MAPK, JNK, NF-κB, TNF-α, and STAT3. Sesquiterpene lactones were shown to be significant in suppressing the proliferation of prostate cancer cell lines (DU-145, PC-3, LNCaP, MR49F, and BPH-1) in both laboratory and clinical settings.
Collapse
Affiliation(s)
- Keshav Kaushal
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, Grand Trunk Rd, Phagwara, Punjab, 144411, India
| | | | - Sanjesh Kumar
- Rakshpal Bahadur College of Pharmacy, Bareilly, U.P, India
| | - Anakha Sony
- Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682041, India
| | - Aswin Viswanath
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, Grand Trunk Rd, Phagwara, Punjab, 144411, India.
| | - M V N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, Grand Trunk Rd, Phagwara, Punjab, 144411, India.
| | - Mansi Singh
- Rakshpal Bahadur College of Pharmacy, Bareilly, U.P, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, Grand Trunk Rd, Phagwara, Punjab, 144411, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology(Pharmacy Institute), 19 Knowledge Park II, Institutional Area, Greater Noida, 201306, India
| |
Collapse
|
17
|
Camacho S, Aoki PHB, Ekstrand F, Oliveira ON, Prinz CN. Enhancing Photothermal Therapy Against Breast Cancer Cells by Modulating the End Point of Gold Shell-Isolated Nanoparticles Using Nanostraw-Assisted Injection. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27816-27828. [PMID: 40299396 PMCID: PMC12086757 DOI: 10.1021/acsami.5c00084] [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] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025]
Abstract
Gold shell-isolated nanoparticles (AuSHINs) are promising photothermal therapy (PTT) agents for cancer treatment due to their excellent photoconversion efficiency, biocompatibility, colloidal stability, and tunable properties, including size, shape, and surface functionalization. However, their therapeutic efficacy in in vitro assays is often limited by poor cellular uptake and lysosomal entrapment, which can result in nanoparticle degradation and a reduction in PTT effectiveness. In this study, we demonstrate that nanostraw-assisted injection enhances the PTT efficacy of AuSHINs compared to the conventional incubation method, as evaluated in human breast cancer cell lines: adenocarcinoma cells (MDA-MB-231) and glandular carcinoma cells (MCF7). This enhancement is attributed to three differences between the delivery methods: nanoparticle internalization, intracellular targeting, and the progression of cell death pathways. Nanostraw injection resulted in approximately 10-fold higher internalization of AuSHINs compared to 0.5-h incubation. Confocal fluorescence microscopy revealed that AuSHINs delivered via conventional incubation predominantly localize within lysosomes, whereas those introduced through nanostraw-assisted injection primarily targeted the endoplasmic reticulum (ER), thus avoiding lysosomal degradation. This differential targeting led to approximately a 2-fold higher reduction in the viability of photoactivated breast cancer cells treated with nanostraw-delivered AuSHINs. Furthermore, nanostraw-assisted injection accelerated the initiation of apoptosis relative to incubation. PTT-induced cell death was more pronounced in MCF7 cells compared to MDA-MB-231 cells, reflecting the higher resistance to therapy of the latter. These findings highlight the potential of nanostraw-assisted injection to enhance PTT, and we now face the challenge of integrating it into in vivo delivery strategies.
Collapse
Affiliation(s)
- Sabrina
A. Camacho
- School
of Sciences, Humanities and Languages, São
Paulo State University (UNESP), Assis, SP 19806-900, Brazil
| | - Pedro H. B. Aoki
- School
of Sciences, Humanities and Languages, São
Paulo State University (UNESP), Assis, SP 19806-900, Brazil
| | - Frida Ekstrand
- Division
of Solid-State Physics and NanoLund, Lund
University, 221 00 Lund, Sweden
| | - Osvaldo N. Oliveira
- São
Carlos Institute of Physics, University
of São Paulo (USP), São Carlos, SP 13566-590, Brazil
| | - Christelle N. Prinz
- Division
of Solid-State Physics and NanoLund, Lund
University, 221 00 Lund, Sweden
| |
Collapse
|
18
|
Wei L, Wang H, Özkan M, Damian-Buda AI, Loynachan CN, Liao S, Stellacci F. Efficient Direct Cytosolic Protein Delivery via Protein-Linker Co-engineering. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27858-27870. [PMID: 40302608 PMCID: PMC12086766 DOI: 10.1021/acsami.5c02360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025]
Abstract
Protein therapeutics have enormous potential for transforming the treatment of intracellular cell disorders, such as genetic disorders and cancers. Due to proteins' cell-membrane impermeability, protein-based drugs against intracellular targets require efficient cytosolic delivery strategies; however, none of the current approaches are optimal. Here, we present a simple approach to render proteins membrane-permeable. We use arginine-mimicking ligand N,N'-dimethyl-1,3-propanediamine (DMPA) to functionalize the surface of a few representative proteins, varying in isoelectric point and molecular weight. We show that when these proteins have a sufficient number of these ligands on their surface, they acquire the property of penetrating the cell cytosol. Uptake experiments at 37 and 4 °C indicate that one of the penetration pathways is energy independent, with no evidence of pore formation, with inhibition assays indicating the presence of other uptake pathways. Functional tests demonstrate that the modified proteins maintain their main cellular function; specifically, modified ovalbumin (OVA) leads to enhanced antigen presentation and modified cytochrome C (Cyto C) leads to enhanced cell apoptosis. We modified bovine serum albumin (BSA) with ligands featuring different hydrophobicity and end group charges and showed that, to confer cytosolic penetration, the ligands must be cationic and that some hydrophobic content improves the penetration efficiency. This study provides a simple strategy for efficiently delivering proteins directly to the cell cytosol and offers important insights into the design and development of arginine-rich cell-penetrating peptide mimetic small molecules for protein transduction.
Collapse
Affiliation(s)
- Lixia Wei
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Heyun Wang
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Melis Özkan
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Andrada-Ioana Damian-Buda
- Institute
of Biomaterials, Department Materials Science and Engineering, Friedrich-Alexander-Universität, Erlangen 91054, Germany
| | - Colleen N. Loynachan
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Suiyang Liao
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Francesco Stellacci
- Institute
of Materials Science and Engineering, École
polytechnique fédérale de Lausanne, Lausanne 1015, Switzerland
- Institute
of Bioengineering, École polytechnique
fédérale de Lausanne, Lausanne 1015, Switzerland
| |
Collapse
|
19
|
Li X, Huang R, Li P, Tang FK, He J, Sun H, Wang X, Wang M, Lan X, Wang X, Wong SSW, Jin L, Leung KCF, Wong HM, Wang S, Guo L, Ding PH, Yu X. Berberine-Functionalized Bismuth-Doped Carbon Dots in a Pathogen-Responsive Hydrogel System: A Multifaceted Approach to Combating Periodontal Diseases. ACS NANO 2025; 19:17554-17577. [PMID: 40313185 PMCID: PMC12080333 DOI: 10.1021/acsnano.5c00561] [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] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
Periodontal disease, a global health burden linked to dysbiotic oral polymicrobial communities and disrupted immune-inflammatory responses, is critically mediated byPorphyromonas gingivalis(Pg)─the keystone pathogen that sabotages host immunity, triggers tissue inflammation and destruction, and disrupts microbiota balance. Effective therapies should combine antimicrobial action, immune modulation, virulence suppression, and microbiome restoration. Bismuth ions and berberine, which exhibit antimicrobial and epithelial barrier-protecting effects, show potential effectiveness in treating periodontal diseases but face practical limitations due to poor water solubility and bioavailability. To address this, we developed bismuth-doped carbon dots functionalized with structure-modified berberine (BiCD-Ber) as a multifunctional nanomedicine. BiCD-Ber eradicated Pg in various forms, restored Pg-perturbed immune responses in gingival fibroblasts, and preserved epithelial barrier integrity. The doped bismuth ions neutralized Pg virulence factors by blocking the catalytic sites of gingipains. To facilitate in vivo delivery, BiCD-Ber was encapsulated in a disulfide-modified hyaluronic acid hydrogel that degrades in response to Pg metabolites. This BiCD-Ber hydrogel system modulated subgingival microbiota, alleviated inflammation in gingiva, and thereby prevented alveolar bone loss. This approach to concurrently eliminating Pg, modulating inflammatory responses , suppressing virulence factors, and restoring microbiota showcases great potential in managing periodontitis effectively.
Collapse
Affiliation(s)
- Xuan Li
- Faculty
of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, PR China
| | - Regina Huang
- Faculty
of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, PR China
| | - Pugeng Li
- Hospital
of Stomatology, Guanghua School of Stomatology, Guangdong Provincial
Key Laboratory of Stomatology, Sun Yat-sen
University, Guangzhou 510055, PR China
| | - Fung Kit Tang
- Faculty
of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, PR China
| | - Jing He
- Faculty
of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, PR China
| | - Hanyu Sun
- Hospital
of Stomatology, Guanghua School of Stomatology, Guangdong Provincial
Key Laboratory of Stomatology, Sun Yat-sen
University, Guangzhou 510055, PR China
| | - Xiaoyu Wang
- Hospital
of Stomatology, Guanghua School of Stomatology, Guangdong Provincial
Key Laboratory of Stomatology, Sun Yat-sen
University, Guangzhou 510055, PR China
| | - Miao Wang
- Faculty
of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, PR China
| | - Xinmiao Lan
- Beijing
Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering
Research Centre of Ministry of Education of China, Beijing Laboratory
of Biomedical Materials, School of Pharmaceutical Science, Capital Medical University, Beijing 100069, PR China
| | - Xinna Wang
- Department
of Mechanical Engineering, The University
of Hong Kong, Hong Kong
SAR 999077, PR China
| | - Sarah Sze Wah Wong
- Immunology
of Fungal Infections Unit, Institut Pasteur, Paris 75015, France
| | - Lijian Jin
- Faculty
of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, PR China
| | - Ken Cham-Fai Leung
- Department
of Chemistry, The Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, PR China
| | - Hai Ming Wong
- Faculty
of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, PR China
| | - Sheng Wang
- State
Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di
Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Lanping Guo
- State
Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di
Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Pei-Hui Ding
- Stomatology
Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310006, PR China
| | - Xiaolin Yu
- Hospital
of Stomatology, Guanghua School of Stomatology, Guangdong Provincial
Key Laboratory of Stomatology, Sun Yat-sen
University, Guangzhou 510055, PR China
| |
Collapse
|
20
|
Cao Y, Yin X, Wu L, Huang D, Wang Z, Wu F, Jiang J, Chen G, Wang Q. High-Efficiency Ocular Delivery of Brain-Derived Neurotrophic Factor and Oligomycin for Neuroprotection in Glaucoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2500623. [PMID: 40357695 DOI: 10.1002/adma.202500623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/19/2025] [Indexed: 05/15/2025]
Abstract
Glaucoma is a retinal neurodegenerative disease characterized by progressive apoptosis of retinal ganglion cells (RGCs) and irreversible visual impairment. Current therapies rarely offer direct protection for RGCs, highlighting the need for new neuroprotective approaches. Although viral delivery of brain-derived neurotrophic factor (BDNF) has shown potential, concerns about retinal inflammation and limited applicability persist. Meanwhile, non-viral vectors remain inefficient for in vivo ocular gene delivery. Here, a highly biocompatible nanoplatform-PBAE-PLGA-Oligomycin-pBDNF nanoparticles (PPOB NPs) is reported-that co-delivers oligomycin (an ATP inhibitor) and a BDNF plasmid to Müller cells in vivo. This nanoplatform attains an unprecedented transfection efficiency of 64.26% in Müller cells, thereby overcoming the limitations of monotherapeutic neurotrophic approaches that fail to inhibit ATP overproduction and attendant inflammatory responses. In a chronic ocular hypertension rat model, oligomycin effectively mitigated RGC damage by suppressing Müller cell hyperactivation and excessive ATP production under elevated intraocular pressure. Concurrently, it synergistically enhanced BDNF expression in Müller cells, achieving robust protection of RGCs and preservation of optic nerve function. These findings underscore the promise of PPOB NPs as a dual-functional platform, featuring high biocompatibility and efficient gene delivery, for multifaceted therapies against glaucoma and other ocular diseases.
Collapse
Affiliation(s)
- Yuheng Cao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xue Yin
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, 21006, China
| | - Lanrong Wu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Dehua Huang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Feng Wu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jiang Jiang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Guangcun Chen
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
21
|
Boix-Montesinos P, Carrascosa-Marco P, Armiñán A, Vicent MJ. Identification of functional biomarkers for personalized nanomedicine in advanced breast cancer in vitro models. J Control Release 2025; 381:113584. [PMID: 40086758 DOI: 10.1016/j.jconrel.2025.113584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/30/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Nanomedicines represent promising advanced therapeutics for the enhanced treatment of breast cancer, the primary cause of cancer-related deaths in women; however, the clinical translation of nanomedicines remains challenging. Advanced in vitro models of breast cancer may improve preclinical evaluations and the identification of biomarkers that aid the stratification of patients who would benefit from a given nanomedicine. In this study, we first developed a matrix-embedded breast cancer cell spheroid model representing the extracellular matrix and confirmed the faithful recapitulation of disease aggressiveness in vitro. We then characterized factors influencing nanomedicine drug release (i.e., cathepsin B levels/activity, reactive oxygen species levels, glutathione levels, and cytoplasmic pH values) and evaluated nanomedicine internalization and cytotoxicity evaluation in our spheroid model. We confirmed the reduced-to-oxidized glutathione ratio as a functional biomarker of disulfide linker-containing polypeptide-drug conjugate effectiveness. We then established a biobank of patient-derived breast cancer organoids that recapitulate clinical intra-tumor and inter-tumor heterogeneity as a more advanced model. Analysis in organoids revealed that patient-specific responses to a polypeptide-based nanomedicine correlated with cathepsin B levels, supporting the potential of the functional biomarker for patient-tailored nanomedicine selection. Our findings highlight that exhaustively characterized advanced in vitro models support the evaluation of nanomedicines and the identification of functional biomarkers.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Paula Carrascosa-Marco
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Ana Armiñán
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCCIII, Madrid, Spain.
| | - María J Vicent
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCCIII, Madrid, Spain.
| |
Collapse
|
22
|
Naz Z, Fareed M, Chaudhary ARH, Snigdha NT, Zafar A, Alsaidan OA, Mangu K, Ahmad S, Aslam M, Rizwanullah M. Exploring the therapeutic potential of ligand-decorated nanostructured lipid carriers for targeted solid tumor therapy. Int J Pharm 2025; 678:125687. [PMID: 40348302 DOI: 10.1016/j.ijpharm.2025.125687] [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: 03/11/2025] [Revised: 04/19/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Solid tumors present significant therapeutic challenges due to their complex pathophysiology, including poor vascularization, dense extracellular matrix, multidrug resistance, and immune evasion. Conventional treatment strategies, such as chemotherapy, radiotherapy, and surgical interventions, are often associated with systemic toxicity, suboptimal drug accumulation at the tumor site, and chemoresistance. Nanostructured lipid carriers (NLCs) have emerged as a promising approach to enhance anticancer therapy. NLCs offer several advantages, including high drug loading capacity, improved bioavailability, controlled release, and enhanced stability. Recent advancements in active targeting strategies have led to the development of ligand-decorated NLCs, which exhibit selective tumor targeting, improved cellular uptake, and reduced systemic toxicity. By functionalizing NLCs with different targeting ligands, site-specific drug delivery can be achieved for better therapeutic efficacy. This review comprehensively explores the potential of ligand-decorated NLCs in solid tumor therapy, highlights their design principles, and mechanisms of tumor targeting. Furthermore, it discusses various receptor-targeted NLCs for the effective treatment of solid tumors. The potential of ligand-decorated NLCs in combination therapy, gene therapy, photothermal therapy, and photodynamic therapy is also explored. Overall, ligand-decorated NLCs represent a versatile and effective strategy to achieve better therapeutic outcomes in solid tumor therapy.
Collapse
Affiliation(s)
- Zrien Naz
- Department of Pharmaceutics, College of Pharmacy, Al Asmarya University, Zliten 218521, Libya
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | | | - Niher Tabassum Snigdha
- Department of Dental Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 602105 Tamil Nadu, India
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka-72341, Al-Jouf, Saudi Arabia
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka-72341, Al-Jouf, Saudi Arabia
| | - Karthik Mangu
- Kogniverse Education and Research, Bionest, Avishkaran (NIPER), Hyderabad-500037, Telangana, India
| | - Shahnawaz Ahmad
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001 Kurdistan Region, Iraq
| | - Md Rizwanullah
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401 Punjab, India.
| |
Collapse
|
23
|
Li R, Hu Y, Hou Y, Li J, Niu X, Wang M, Wang L. Receptor-mediated membrane fusion drug delivery system based on chitosan derivatives to enhance tumor chemotherapy. Int J Biol Macromol 2025; 311:143869. [PMID: 40348215 DOI: 10.1016/j.ijbiomac.2025.143869] [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: 01/04/2025] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
Tumor chemotherapy drug delivery systems often face significant challenges, including low targeting specificity and lysosomal sequestration, both of which can severely impair therapeutic efficacy. To overcome these limitations, we have developed a novel receptor-mediated membrane fusion (RMF) drug delivery system based on chitosan derivatives. This system can self-assemble into nanoparticles (NPs) and encapsulate doxorubicin (DOX). The physical properties of both unloaded and DOX-loaded NPs were systematically characterized. In vitro experiments demonstrated that the RMF system selectively interacts with tumor cell surfaces, inhibiting cell proliferation and migration. Additionally, the system effectively targets tumor cells, delivers the drug directly into the cytoplasm, thereby bypassing lysosomal sequestration, thus improving targeting efficiency and enhancing drug delivery. In vivo studies further confirmed the superior anticancer efficacy of the RMF system, alongside its excellent systemic safety. In conclusion, this RMF-based strategy offers a promising platform for the precise delivery of chemotherapeutics, addressing the critical limitations of conventional drug delivery systems and significantly enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Ruxiang Li
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Yaqi Hu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Yiyang Hou
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Jingge Li
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Xiaoyuan Niu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Mandi Wang
- Department of Chemistry, University of Wisconsin-Madison, 500 Lincoln Avenue, 161 Bascom Hall, Madison 53706, WI, United States.
| | - Lianyong Wang
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China.
| |
Collapse
|
24
|
Mendez-Pfeiffer P, Ballesteros Monrreal MG, Mendez-Encinas MA, Valencia D, Ortiz B, González-Davis O, Cadena-Nava RD. Nanoparticles in Antibacterial Therapy: A Systematic Review of Enhanced Efficacy against Intracellular Bacteria. ACS OMEGA 2025; 10:17070-17086. [PMID: 40352514 PMCID: PMC12059930 DOI: 10.1021/acsomega.5c01813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/01/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
Intracellular bacterial infections represent a considerable therapeutic challenge due to the ability of pathogens to invade and replicate within host cells, hampering the action of the immune system and the effectiveness of conventional antibiotics. Bacteria such as Mycobacterium tuberculosis, Listeria monocytogenes, and methicillin-resistant Staphylococcus aureus (MRSA), among others, can persist within host cells, allowing them to evade the immune response and develop resistance to antibacterial treatments. A key factor in the persistence of these infections is the ability of bacteria to enter a dormant state, which reduces their susceptibility to antibiotics that affect the dividing cells. Nanotechnology is emerging as a promising solution as nanoparticle-based systems can improve the intracellular penetration of antibiotics, allow their controlled release, and reduce side effects. This review covers the development and efficacy of nanoparticle-encapsulated antibiotics in models of intracellular infections, highlighting the need to further investigate their potential to overcome the barriers of conventional therapies and improve the treatment of these complex infections.
Collapse
Affiliation(s)
- Pablo Mendez-Pfeiffer
- Departamento
de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera Tijuana-Ensenada, Ensenada, Baja California 22860, México
- Departamento
de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Campus Caborca, Caborca 83600, Sonora, México
| | - Manuel G. Ballesteros Monrreal
- Departamento
de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Campus Caborca, Caborca 83600, Sonora, México
| | - Mayra A. Mendez-Encinas
- Departamento
de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Campus Caborca, Caborca 83600, Sonora, México
| | - Dora Valencia
- Departamento
de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Campus Caborca, Caborca 83600, Sonora, México
| | - Bryan Ortiz
- Instituto
de Investigaciones en Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa 11101, Honduras
| | - Oscar González-Davis
- Departamento
de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera Tijuana-Ensenada, Ensenada, Baja California 22860, México
| | - Ruben D. Cadena-Nava
- Departamento
de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km 107 Carretera Tijuana-Ensenada, Ensenada, Baja California 22860, México
| |
Collapse
|
25
|
Liang M, Zhou L, Li J, Liang B, Zhou L, Xue F, Jiang L, Hong W. An acid-responsive bone-targeting nanoplatform loaded with curcumin balances osteogenic and osteoclastic functions. Regen Biomater 2025; 12:rbaf028. [PMID: 40443874 PMCID: PMC12122077 DOI: 10.1093/rb/rbaf028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/20/2025] [Accepted: 04/13/2025] [Indexed: 06/02/2025] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a predominant form of clinical osteoporosis. It has led to significant health and social burdens for older patients. Reestablishing the balance between osteogenic and osteoclastic is a crucial strategy for treating PMOP. Curcumin (Cur), a naturally derived polyphenolic substance, has gained recognition as a viable option for treating osteoporosis. Despite its potential, the clinical use of Cur is hindered by its limited bioavailability and the presence of side effects. Nanoparticles modified with aspartic acid octapeptide (ASP8) exhibit a strong affinity for bone tissue, facilitating targeted delivery. This study presents novel acid-responsive zeolite imidazolate framework-8 (ZIF) nanoparticles modified with ASP8 and loaded with Cur (Cur@ZIF@ASP8, CZA). Upon delivery by this nanoparticle drug delivery system, Cur can effectively regulate bone homeostasis, offering a potential therapeutic strategy for osteoporosis. This study demonstrated that CZA nanoparticles could successfully transport Cur to bone tissue without significant toxicity. Furthermore, nanoparticles promote bone formation and inhibit osteoclast activity. They also modify the expression of related genes and proteins, such as OCN, ALP, CTSK and MMP9. Significant evaluations utilizing microcomputed tomography, Masson's staining, hematoxylin and eosin staining and immunofluorescence staining demonstrated that intravenous CZA administration in ovariectomized mice resulted in bone destruction while simultaneously reducing overall bone loss. In conclusion, CZA nanoparticles hold promise as a therapeutic option for osteoporosis.
Collapse
Affiliation(s)
- Minhao Liang
- Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital, Fudan University, Shanghai 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Lei Zhou
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Juan Li
- Department of Orthopaedic Surgery, Shanghai Geriatric Medical Center, Shanghai 201104, China
| | - Bin Liang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liangyun Zhou
- Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital, Fudan University, Shanghai 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Fengfeng Xue
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200070, China
| | - Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Orthopedic Surgery, Zhongshan Hospital (Xiamen Branch), Fudan University, Shanghai 200032, China
| | - Wei Hong
- Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital, Fudan University, Shanghai 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
26
|
Salehi Moghaddam A, Bahrami M, Sarikhani E, Tutar R, Ertas YN, Tamimi F, Hedayatnia A, Jugie C, Savoji H, Qureshi AT, Rizwan M, Maduka CV, Ashammakhi N. Engineering the Immune Response to Biomaterials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414724. [PMID: 40232044 PMCID: PMC12097135 DOI: 10.1002/advs.202414724] [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] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/29/2025] [Indexed: 04/16/2025]
Abstract
Biomaterials are increasingly used as implants in the body, but they often elicit tissue reactions due to the immune system recognizing them as foreign bodies. These reactions typically involve the activation of innate immunity and the initiation of an inflammatory response, which can persist as chronic inflammation, causing implant failure. To reduce these risks, various strategies have been developed to modify the material composition, surface characteristics, or mechanical properties of biomaterials. Moreover, bioactive materials have emerged as a new class of biomaterials that can induce desirable tissue responses and form a strong bond between the implant and the host tissue. In recent years, different immunomodulatory strategies have been incorporated into biomaterials as drug delivery systems. Furthermore, more advanced molecule and cell-based immunomodulators have been developed and integrated with biomaterials. These emerging strategies will enable better control of the immune response to biomaterials and improve the function and longevity of implants and, ultimately, the outcome of biomaterial-based therapies.
Collapse
Affiliation(s)
- Abolfazl Salehi Moghaddam
- Department of BioengineeringP.C. Rossin College of Engineering & Applied ScienceLehigh UniversityBethlehemPA18015USA
| | - Mehran Bahrami
- Department of Mechanical Engineering & MechanicsLehigh UniversityBethlehemPA18015USA
| | - Einollah Sarikhani
- Department of Nano and Chemical EngineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Rumeysa Tutar
- Department of ChemistryFaculty of Engineering, Istanbul University‐CerrahpaşaIstanbul, Avcılar34320Turkey
| | - Yavuz Nuri Ertas
- Department of Biomedical EngineeringErciyes UniversityKayseri38039Turkey
- ERNAM – Nanotechnology Research and Application CenterErciyes UniversityKayseri38039Turkey
| | - Faleh Tamimi
- College of Dental MedicineQatar University HealthQatar UniversityP.O. Box 2713DohaQatar
| | - Ali Hedayatnia
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Institute of Biomedical Engineering, Department of Pharmacology and PhysiologyFaculty of MedicineMontrealQuebecH3T 1J4Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Clotilde Jugie
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Houman Savoji
- Azrieli Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Institute of Biomedical Engineering, Department of Pharmacology and PhysiologyFaculty of MedicineMontrealQuebecH3T 1J4Canada
- Montreal TransMedTech InstituteiTMTMontrealQuebecH3T 1J4Canada
| | - Asma Talib Qureshi
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
| | - Muhammad Rizwan
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
- Health Research InstituteMichigan Technological UniversityHoughtonMI49931USA
| | - Chima V. Maduka
- BioFrontiers InstituteUniversity of ColoradoBoulderCO80303USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Colleges of Engineering and Human MedicineMichigan State UniversityEast LansingMI48824USA
- Department of BioengineeringSamueli School of EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
27
|
Li CS, Liu J, Zhang Q, Tang XR, Liu YY, Cao A, Wang H. Combined Effect of Nanoparticles of Silver and Silica to HeLa Cells: Synergistic Internalization and Toxicity. ENVIRONMENTAL TOXICOLOGY 2025; 40:802-816. [PMID: 39840792 DOI: 10.1002/tox.24480] [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: 06/11/2024] [Revised: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025]
Abstract
The wide range of applications and the enormous production of nanomaterials have raised the possibility that humans may simultaneously contact with various nanomaterials through multiple routes. Although numerous toxicity studies have been conducted on the toxicity of nanomaterials, knowledge of the combined toxicity of nanomaterials remains limited. Herein, the combined toxic effects of the two types of the most widely used nanomaterials, silver and silica, were studied on HeLa cells. In addition, considering there may have possible interplay between nanoparticles of different sizes, two different-sized silica nanoparticles (SNPs) were used. The results indicate that compared with individual exposure, the combined exposure to 35 nm silver nanoparticles (Ag35) and 40 nm or 120 nm SNPs (SNP40 or SNP120) at individual non-toxic concentrations causes more severe cytotoxicity, manifested by the ROS overgeneration, decreased mitochondrial membrane potential and ATP level, and increased apoptosis/necrosis. The internalized Ag35 and its dissolved Ag ions that are delivered into cells by adsorbing on SNPs are identified as the primary contributors to the combined toxicity. Although the cytotoxicity of the mixed Ag35 and SNP40 is comparable to that of the mixed Ag35 and SNP120, there are noticeable differences in their intracellular contents and their subcellular locations due to size effects. This study provides in-depth insights into the combined toxicity of inorganic nanoparticles and highlights the importance of the size effect of nanoparticles in their nanotoxicity assessment.
Collapse
Affiliation(s)
- Chen-Si Li
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Jie Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Qiangqiang Zhang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Xue-Rui Tang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Yuan-Yuan Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Aoneng Cao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| |
Collapse
|
28
|
Maeda N, Jiao H, Kłosowska‐Chomiczewska IE, Artichowicz W, Preiss U, Szumała P, Macierzanka A, Jungnickel C. Nanoparticle Skin Penetration: Depths and Routes Modeled In-Silico. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412541. [PMID: 40150997 PMCID: PMC12087856 DOI: 10.1002/smll.202412541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Nanoparticles (NPs) are increasingly explored for targeted skin penetration, particularly for pharmaceutical and cosmetic applications. However, the complex system between NP properties, skin structure, and experimental conditions poses significant challenges in predicting their penetration depth and pathways. To what depth do NPs penetrate the skin, and which pathways do they follow? These are the questions which we tried to answer in this paper. A n in-silico human skin model based on 20 years of literature on NPs skin penetration is developed. The model incorporates 19 independent parameters, including a wide range of NP properties, skin across species, and test conditions. Using random forest analysis coupled with Kennard-Stone sorting, the model achieves a high predictive accuracy of 95%. The study identifies hair follicle diameter as the most critical factor influencing NP penetration across skin layers, surpassing other skin properties, NP properties, or experimental variables. Pig and rabbit skin are the most suitable models for simulating human skin in NP penetration studies. Additionally, the in-silico model reveals that NPs in emulsions and oil-based media predominantly follow the intercellular and transappendageal route. In contrast, those embedded in aqueous media favor the intracellular route. These findings offer insights for optimizing NP-based drug delivery systems.
Collapse
Affiliation(s)
- Natsumi Maeda
- Institute of Biogeochemistry and Pollutant DynamicsSwiss Federal Institute of TechnologyETH ZürichUniversitätstrasse 16Zürich8092Switzerland
| | - Haixin Jiao
- Biofuels InstituteSchool of the Environment and Safety EngineeringJiangsu UniversityZhenjiang212013P. R. China
| | | | - Wojciech Artichowicz
- Department of Hydraulic EngineeringFaculty of Civil and Environmental EngineeringGdańsk University of TechnologyNarutowicza 11/12Gdańsk80–233Poland
| | - Ulrich Preiss
- SustainabilityOmya GmbHSiegburger Str. 229c50679KölnGermany
| | - Patrycja Szumała
- Department of Biotechnology and MicrobiologyFaculty of ChemistryGdańsk University of TechnologyNarutowicza 11/12Gdańsk80–233Poland
| | - Adam Macierzanka
- Department of Biotechnology and MicrobiologyFaculty of ChemistryGdańsk University of TechnologyNarutowicza 11/12Gdańsk80–233Poland
| | - Christian Jungnickel
- Department of Biotechnology and MicrobiologyFaculty of ChemistryGdańsk University of TechnologyNarutowicza 11/12Gdańsk80–233Poland
| |
Collapse
|
29
|
Song S, Han H, Wang J, Pu Y, Shao J, Xie J, Che H, van Hest JCM, Cao S. Polymersome-based nanomotors: preparation, motion control, and biomedical applications. Chem Sci 2025; 16:7106-7129. [PMID: 40206551 PMCID: PMC11976864 DOI: 10.1039/d4sc08283d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
Polymersome-based nanomotors represent a cutting-edge development in nanomedicine, merging the unique vesicular properties of polymersomes with the active propulsion capabilities of synthetic nanomotors. As a vesicular structure enclosed by a bilayer membrane, polymersomes can encapsulate both hydrophilic and hydrophobic cargoes. In addition, their physical-chemical properties such as size, morphology, and surface chemistry are highly tunable, which makes them ideal for various biomedical applications. The integration of motility into polymersomes enables them to actively navigate biological environments and overcome physiological barriers, offering significant advantages over passive delivery platforms. Recent breakthroughs in fabrication techniques and motion control strategies, including chemically, enzymatically, and externally driven propulsion, have expanded their potential for drug delivery, biosensing, and therapeutic interventions. Despite these advancements, key challenges remain in optimizing propulsion efficiency, biocompatibility, and in vivo stability to translate these systems into clinical applications. In this perspective, we discuss recent advancements in the preparation and motion control strategies of polymersome-based nanomotors, as well as their biomedical-related applications. The molecular design, fabrication approaches, and nanomedicine-related utilities of polymersome-based nanomotors are highlighted, to envisage the future research directions and further development of these systems into effective, precise, and smart nanomedicines capable of addressing critical biomedical challenges.
Collapse
Affiliation(s)
- Siyu Song
- Life-Like Materials and Systems, Department of Chemistry, University of Mainz Mainz 55128 Germany
| | - Hao Han
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 PR China
| | - Jianhong Wang
- Bio-Organic Chemistry, Institute of Complex Molecular Systems, Eindhoven University of Technology Helix, P. O. Box 513 Eindhoven 5600 MB The Netherlands
| | - Yubin Pu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 PR China
| | - Jingxin Shao
- Bio-Organic Chemistry, Institute of Complex Molecular Systems, Eindhoven University of Technology Helix, P. O. Box 513 Eindhoven 5600 MB The Netherlands
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University Chengdu 610041 China
| | - Hailong Che
- Department of Chemical Engineering, School of Environmental and Chemical Engineering, Shanghai University Shanghai 200444 China
| | - Jan C M van Hest
- Bio-Organic Chemistry, Institute of Complex Molecular Systems, Eindhoven University of Technology Helix, P. O. Box 513 Eindhoven 5600 MB The Netherlands
| | - Shoupeng Cao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 PR China
| |
Collapse
|
30
|
Di Stolfo L, Lee WS, Vanhecke D, Balog S, Taladriz-Blanco P, Petri-Fink A, Rothen-Rutishauser B. The impact of cell density variations on nanoparticle uptake across bioprinted A549 gradients. Front Bioeng Biotechnol 2025; 13:1584635. [PMID: 40370598 PMCID: PMC12075422 DOI: 10.3389/fbioe.2025.1584635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction The safe-by-design of engineered nanoparticles (NPs) for any application requires a detailed understanding of how the particles interact with single cells. Most studies are based on two-dimensional, uniformly dense cell cultures, which do not represent the diverse and inhomogeneous cell environments found in situ. In-vitro models that accurately represent tissue complexity, including realistic cell densities, are essential to increase the predictive accuracy of studies on cell-NP interactions. This study uses a bioprinted cell gradient model to examine the relation between cell density and NP uptake in one dish. Method A549 lung epithelial cell density gradients within single inserts were produced with a bioprinter by modulating inter-droplet distances. After two days in culture, cells were exposed to Cy5-labeled silica NPs (SiO2 NPs, ∼112 nm, 20 μg/mL) for up to 48 h. Confocal fluorescence microscopy and 3D image analysis were used to quantify NP uptake, cell surface area, and cell volume. The relationship between NP uptake and the other parameters was then investigated statistically. Results Bioprinting enabled the creation of reproducible linear cell density gradients, allowing controlled modeling of density variations while preserving cell viability throughout the experiment. Increasing inter-droplet distances, from 0.1 mm to 0.6 mm, were used to achieve uniformly decreasing cell densities. SiO2 NP uptake per cell was around 50% higher in low-density regions compared to high-density areas across all time points, i.e., 6, 24, and 48 h post-exposure. This inverse relationship correlated with greater average cell surface area in lower-density regions, while differences in the proliferation rates of the A549 cells at varying densities did not significantly impact uptake, did not significantly impact uptake. Conclusion SiO2 NP uptake is significantly enhanced at lower cell densities, mainly due to the increased available surface area, revealing potential cell-NP interaction differences in tissues that present cell density variability. Our drop-on-demand bioprinting gradient model successfully supports the implementation of cell density gradients in in-vitro models to increase their relevance as new approach methodologies (NAMs) for next-generation risk assessment strategies.
Collapse
Affiliation(s)
- Luigi Di Stolfo
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Wang Sik Lee
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Dimitri Vanhecke
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Sandor Balog
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Patricia Taladriz-Blanco
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Alke Petri-Fink
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
- Department of Chemistry, University of Fribourg, Fribourg, Switzerland
| | - Barbara Rothen-Rutishauser
- Adolphe Merkle Institute and National Center of Competence in Research Bio-Inspired Materials, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
31
|
Lai S, Tang N, Guo J, Deng L, Yuan L, Zeng L, Yang L, Mu Y. Immunomodulatory peptide DP7-C mediates macrophage-derived exosomal miR-21b to promote bone regeneration via the SOCS1/JAK2/STAT3 axis. Colloids Surf B Biointerfaces 2025; 253:114709. [PMID: 40286607 DOI: 10.1016/j.colsurfb.2025.114709] [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: 11/03/2024] [Revised: 03/18/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
Periodontitis, the most prevalent chronic inflammatory disease leading to bone resorption, presents significant challenges for achieving optimal periodontal bone regeneration and repair despite efforts to reduce inflammation and stimulate osteogenesis. Macrophage-derived exosomes have emerged as promising therapeutic agents due to their osteogenic and immunomodulatory potential. Specific stimulation of macrophages can alter the exosomal composition, particularly microRNAs (miRNAs), thereby altering their functions. DP7-C, a cationic immunomodulatory peptide, is known to regulate immune responses and cellular processes by interacting with cell membranes and signaling pathways. However, its effects on macrophage exosomal miRNA profiles remain poorly understood. In this study, we identified differential miRNA expression in macrophage-derived exosomes following DP7-C stimulation, with a notable upregulation of miR-21b. To investigate the osteogenic role of exosomal miR-21b, DP7-C was utilized to facilitate the transfection of miR-21b into macrophages, leading to the secretion of exosomes enriched with miR-21b. These exosomes enhanced osteogenic differentiation in vitro and alleviated periodontal tissue damage in an experimental periodontitis model in vivo. Mechanistically, exosomal miR-21b promotes osteogenesis by directly targeting the suppressor of cytokine signaling (SOCS1), thereby activating the JAK2/STAT3 signaling pathway. This study establishes macrophage-derived exosomal miR-21b as a potent catalyst for bone regeneration, highlighting a promising acellular therapeutic strategy for periodontitis.
Collapse
Affiliation(s)
- Shuang Lai
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Na Tang
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Jun Guo
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Li Deng
- Centerfor Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan 611135, China
| | - Lun Yuan
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Linya Zeng
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Lu Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Yandong Mu
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| |
Collapse
|
32
|
Yang YP, Nicol CJB, Chiang MC. A Review of the Neuroprotective Properties of Exosomes Derived from Stem Cells and Exosome-Coated Nanoparticles for Treating Neurodegenerative Diseases and Stroke. Int J Mol Sci 2025; 26:3915. [PMID: 40332773 PMCID: PMC12028030 DOI: 10.3390/ijms26083915] [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: 02/18/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Neurological diseases, including neurodegenerative disorders and stroke, represent significant medical challenges due to their complexity and the limitations of current treatment approaches. This review explores the potential of stem cell (SC)-derived exosomes (Exos) as a transformative therapeutic strategy for these diseases. Exos, especially those derived from SCs, exhibit natural targeting ability, biocompatibility, and the capacity to cross the blood-brain barrier (BBB), making them ideal vehicles for drug delivery. This review provides an in-depth discussion of the properties and advantages of SC-Exos. It highlights their potential synergistic benefits in therapeutic approaches to treat neurological diseases. This article discusses the mechanisms of action of SC-Exos, highlighting their ability to target specific cells, modulate disease pathways, and provide controlled release of therapeutic agents. Applications in specific neurological disorders have been investigated, demonstrating the potential to improve outcomes in conditions such as Alzheimer's Disease (AD), Parkinson's Disease (PD), and stroke. Moreover, Exos-coated nanoparticles (NPs) combine the natural properties of Exos with the multifunctionality of NPs. This integration takes advantage of exosome membrane biocompatibility and targeting capabilities while preserving NPs' beneficial features, such as drug loading and controlled release. As a result, Exos-coated NPs may enhance the precision, efficacy, and safety of therapeutic interventions. In conclusion, SC-Exos represent a promising and innovative approach to treating neurological diseases.
Collapse
Affiliation(s)
- Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, and Cancer Biology and Genetics Division, Sinclair Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
33
|
Zhao J, Duan Z, Chen Y, Wu Q, Xie J, Si J, Wang Y, Xiang J, Shao S, Shen Y. Zwitterionic Internalizable Peptide-Drug Conjugates: Tumor-Selective Ultrafast Uptake and Transcytosis for Enhanced Antitumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501485. [PMID: 40227128 DOI: 10.1002/smll.202501485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/19/2025] [Indexed: 04/15/2025]
Abstract
Here a novel class of zwitterionic internalizable peptide (ZIP)-drug conjugates is presented. The conjugates incorporate γ-glutamyl-α-aminobutyrylamino-functionalized lysine peptides as the ZIP backbones and disulfide-linked camptothecin (CPT) as the toxic payload. The ZIPs significantly enhance the water solubility of CPT without compromising its cytotoxicity. Cellular uptake profiling shows ultrafast internalization of the conjugates in tumor cells within 5 min. Mechanistic studies reveal that they enter cells via lipid raft-mediated pathways, involving both energy-driven active transport and membrane fluidity-dependent passive uptake. Furthermore, these conjugates exhibit robust transcytosis abilities across dense cell monolayers and tumor spheroids. These characteristics confer prolonged blood circulation, enhanced tumor accumulation, and deep tumor penetration on the conjugates, which result in potent in vivo antitumor activity and minimal systemic toxicity. This study underscores the potential of the conjugates for safer and more effective cancer therapies and presents ZIPs as a new category of molecularly defined delivery tools for biomedical applications.
Collapse
Affiliation(s)
- Jianglin Zhao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zhongkun Duan
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yuping Chen
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Qi Wu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jingwen Xie
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jingxing Si
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yi Wang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
34
|
Soh WWM, Zhu J, Zhang Z, Mazlan MDM, Chin EWM, Cheah CH, Goh ELK, Li J. Supramolecular Polycations with a Linear-Star Architecture Containing Hydrophobic Poly[( R, S)-3-hydroxybutyrate]: Formation of DNA Micelleplexes Coated with Apolipoprotein E3 for Blood-Brain Barrier Penetrating Gene Delivery. Biomacromolecules 2025; 26:2157-2170. [PMID: 40052737 DOI: 10.1021/acs.biomac.4c01412] [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: 04/15/2025]
Abstract
A novel blood-brain barrier (BBB)-penetrating supramolecular gene delivery system was developed utilizing a host-guest block-building strategy to systematically screen and optimize various block compositions. Linear poly(ethylene glycol) (PEG) was coupled with hydrophobic poly[(R,S)-β-hydroxybutyrate] (PHB) blocks of varying lengths with an adamantyl (Ad) end, giving the PEG-PHB-Ad guest polymers, which were complexed with the cationic 4-arm star-shaped β-cyclodextrin-poly(2-dimethylaminoethyl methacrylate) (βCD-pDMAEMA) host polymer, resulting in the formation of linear-star pseudoblock PEG-PHB-Ad/βCD-pDMAEMA copolymers. These amphiphilic supramolecular copolymers were thoroughly characterized and assessed for the formation of DNA micelleplex nanoparticles as a gene delivery system. Through a rational selection process, an optimal host-guest configuration was identified, considering critical factors such as cytotoxicity, gene transfection efficiency, serum stability, cellular uptake, and hemolytic activity. The optimized host-guest copolymer was subsequently coated with the targeting protein apolipoprotein E3 (ApoE3), endowing it with BBB-penetrating capabilities, which was validated through an in vitro BBB transwell model.
Collapse
Affiliation(s)
- Wilson Wee Mia Soh
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Zhongxing Zhang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Muhammad Danial Mohd Mazlan
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eunice W M Chin
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Chee Hoe Cheah
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eyleen L K Goh
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Jun Li
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
- NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
35
|
Wong CN, Lee SK, Lim YM, Yang SB, Chew YL, Chua AL, Liew KB. Recent Advances in Vitamin E TPGS-Based Organic Nanocarriers for Enhancing the Oral Bioavailability of Active Compounds: A Systematic Review. Pharmaceutics 2025; 17:485. [PMID: 40284480 PMCID: PMC12030195 DOI: 10.3390/pharmaceutics17040485] [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: 02/28/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Background: D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS), an amphiphilic derivative of natural vitamin E, functions as both a drug efflux inhibitor and a protector against enzymatic degradation and has been widely incorporated into nano-formulations for drug design and delivery. Objective: This systematic review evaluates TPGS-based organic nanocarriers, emphasizing their potential to enhance bioavailability of active compounds which include drugs and phytochemicals, improve pharmacokinetic profiles, and optimize therapeutic outcomes, eventually overcoming the limitations of conventional oral active compounds delivery. Search strategy: Data collection was carried out by entering key terms (TPGS) AND (Micelle OR Liposome OR Nanoparticle OR Nanotube OR Dendrimer OR Niosome OR Nanosuspension OR Nanomicelle OR Nanocrystal OR Nanosphere OR Nanocapsule) AND (Oral Bioavailability) into the Scopus database. Inclusion criteria: Full-text articles published in English and relevant to TPGS, which featured organic materials, utilized an oral administration route, and included pharmacokinetic study, were included to the final review. Data extraction and analysis: Data selection was conducted by two review authors and subsequently approved by all other authors through a consensus process. The outcomes of the included studies were reviewed and categorized based on the types of nanocarriers. Results: An initial search of the database yielded 173 records. After screening by title and abstract, 52 full-text articles were analyzed. A total of 21 papers were excluded while 31 papers were used in this review. Conclusions: This review concludes that TPGS-based organic nanocarriers are able to enhance the bioavailability of various active compounds, including several phytochemicals, leveraging TPGS's amphiphilic nature, inhibition of efflux transporters, protection against degradation, and stabilization properties. Despite using the same excipient, variability in particle size, zeta potential, and encapsulation efficiency among nanocarriers indicates the need for tailored formulations. A comprehensive approach involving the development and standardized comparison of diverse TPGS-incorporated active compound formulations is essential to identify the optimal TPGS-based nanocarrier for improving a particular active compound's bioavailability.
Collapse
Affiliation(s)
- Chee Ning Wong
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia; (C.N.W.); (Y.M.L.)
| | - Siew-Keah Lee
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia; (C.N.W.); (Y.M.L.)
| | - Yang Mooi Lim
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia; (C.N.W.); (Y.M.L.)
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan;
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia;
| | - Ang-Lim Chua
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Malaysia;
| | - Kai Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Cyberjaya 63000, Malaysia
| |
Collapse
|
36
|
Nordin NA, Sadikan MZ, Lambuk L, Hashim S, Airuddin S, Mohd Nasir NA, Mohamud R, Ibrahim J, Kadir R. Liposomal topical drug administration surpasses alternative methods in glaucoma therapeutics: a novel paradigm for enhanced treatment. J Pharm Pharmacol 2025; 77:475-491. [PMID: 39579384 DOI: 10.1093/jpp/rgae129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/01/2024] [Indexed: 11/25/2024]
Abstract
OBJECTIVES Glaucoma is a leading cause of permanent blindness. Despite therapeutic advancements, glaucoma management remains challenging due to limitations of conventional drug delivery, primarily topical eye drops, resulting in suboptimal outcomes and a global surge in cases. To address these issues, liposomal drug delivery has emerged as a promising approach. KEY FINDINGS This review explores the potential of liposomal-based medications, with a particular focus on topical administration as a superior alternative to enhance therapeutic efficacy and improve patient compliance compared to existing treatments. This writing delves into the therapeutic prospects of liposomal formulations across different administration routes, as evidenced by ongoing clinical trials. Additionally, critical aspects of liposomal production and market strategies are discussed herein. SUMMARY By overcoming ocular barriers and optimizing drug delivery, liposomal topical administration holds the key to significantly improving glaucoma treatment outcomes.
Collapse
Affiliation(s)
- Nor Asyikin Nordin
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Muhammad Zulfiqah Sadikan
- Department of Pharmacology, Faculty of Medicine, Manipal University College Malaysia (MUCM), 75150 Bukit Baru, Melaka, Malaysia
| | - Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Sabarisah Hashim
- Department of Neurosciences, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Malaysia
| | - Syahira Airuddin
- Reconstructive Science Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Nur-Azida Mohd Nasir
- Reconstructive Science Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Jamal Ibrahim
- Maths, Science and IT Curriculum Area, Oxford Sixth Form College, 12-13 King Edward St, Oxford, OX1 4HT, United Kingdom
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
37
|
Shafiee M, Soltani Fard E, Taghvimi S, Movahedpour A, Mousavi P, Rezaeijo SM, Khatami SH, Azadbakht O. "Nanoparticle-based sensitizers in prostate cancer treatment: Enhancing radiotherapy efficacy through innovative nanotechnology: Narrative review". Appl Radiat Isot 2025; 218:111671. [PMID: 39826201 DOI: 10.1016/j.apradiso.2025.111671] [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: 05/29/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
For men with localized prostate cancer, radiotherapy (RT) remains a common therapeutic option. Although radiotherapy has had significant success, it remains an intractable issue in promoting radiation damage to tumor tissue while reducing adverse effects on healthy tissue. Chemicals or pharmacological substances known as radiosensitizers can increase the killing effect on tumor cells by accelerating DNA damage and indirectly producing free radicals. Of all the approaches to improving RT management outcomes, metal nanoparticle-enhanced radiation for prostate cancer patient therapy is a unique strategy that has sparked scientific attention in the past decade. Most current data is based on targeted RT with gold nanoparticles, among the most studied materials. Nevertheless, several novel materials have also been employed in preclinical settings. This study assesses existing dosimetric data on prostate cancer tissue as well as the likely future influence on treatment options and patient outcomes since further research in a clinical setting is necessary.
Collapse
Affiliation(s)
- Mohsen Shafiee
- Department of Nursing, Abadan University of Medical Sciences, Abadan, Iran.
| | - Elahe Soltani Fard
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Sina Taghvimi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | | | - Pegah Mousavi
- Molecular Medicine Research Center, Hormozgan Health Institude, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Seyed Masoud Rezaeijo
- Department of Medical Physics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
38
|
Chen X, Dai W, Liang W, Li D, Cai WQ, Zhang C, Hu W, Zhang BB, Yang Q. ROS-responsive nanocarrier for oral delivery of monascin and enhanced alleviation of oxidative stress. J Biosci Bioeng 2025; 139:319-328. [PMID: 39864970 DOI: 10.1016/j.jbiosc.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/12/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025]
Abstract
Oxidative stress, caused by excessive production of reactive oxygen species (ROS), plays a crucial role in the occurrence and development of various diseases. Monascin can scavenge ROS and alleviate oxidative stress but with a low fermentation rate and bioavailability. Here, we optimized the fermentation process to increase the production of monascin (508.6 U/mL), and then systematically characterized its structure via HPLC, HPLC-MS, 1H NMR, and 13C NMR. Additionally, we innovatively modified carboxymethylcellulose sodium with selenium (CMC-Se) to encapsulate monascin (monascin@CMC-Se), which can sensitively respond to ROS and release monascin to effectively scavenge excessive ROS. Besides, the monascin@CMC-Se can significantly increase the bioaccessibility of monascin and alleviate cellular oxidative stress by enhancing its cellular uptake rate. Collectively, our work provides proof-of-principle evidence that the CMC-Se can precise delivery of monascin to an oxidatively stressed environment with high resistance to gastric fluids, laying a foundation to overcome inflammation-related diseases in the colon.
Collapse
Affiliation(s)
- Xueyi Chen
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; Shantou Key Laboratory of Marine Microbial Resources and Interactions with Environment, Shantou University, Shantou 515063, China
| | - Wenli Dai
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; Shantou Key Laboratory of Marine Microbial Resources and Interactions with Environment, Shantou University, Shantou 515063, China
| | - Wanqi Liang
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; Shantou Key Laboratory of Marine Microbial Resources and Interactions with Environment, Shantou University, Shantou 515063, China
| | - Dong Li
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou 515031, China
| | - Wo-Qi Cai
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; Shantou Key Laboratory of Marine Microbial Resources and Interactions with Environment, Shantou University, Shantou 515063, China
| | - Chan Zhang
- School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Wenlin Hu
- Guangdong Tianyi Biotechnology Co., Ltd., Zhanjiang 524000, China
| | - Bo-Bo Zhang
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; Shantou Key Laboratory of Marine Microbial Resources and Interactions with Environment, Shantou University, Shantou 515063, China.
| | - Qiongqiong Yang
- Department of Biology, College of Science, Shantou University, Shantou 515063, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China; Shantou Key Laboratory of Marine Microbial Resources and Interactions with Environment, Shantou University, Shantou 515063, China.
| |
Collapse
|
39
|
Atchudan R, Karuppasamy BD, Perumal S, Gangadaran P, Sundramoorthy AK, Manoj D, Rajendran RL, Ahn BC, Ahamed M, Lee SW, Lee YR. Sustainable-biomass-derived multifunctional carbon dots as fluorescent probes for multi-purpose advanced imaging, migration and security solutions. SURFACES AND INTERFACES 2025; 62:106238. [DOI: 10.1016/j.surfin.2025.106238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
|
40
|
Neupane R, Malla S, Karthikeyan C, Asbhy CR, Boddu SHS, Jayachandra Babu R, Tiwari AK. Endocytic highways: Navigating macropinocytosis and other endocytic routes for precision drug delivery. Int J Pharm 2025; 673:125356. [PMID: 39956408 DOI: 10.1016/j.ijpharm.2025.125356] [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: 09/15/2024] [Revised: 12/22/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Drug molecules can reach intracellular targets by different mechanisms, such as passive diffusion, active transport, and endocytosis. Endocytosis is the process by which cells engulf extracellular material by forming a vesicle and transporting it into the cells. In addition to its biological functions, endocytosis plays a vital role in the internalization of the therapeutic molecules. Clathrin-mediated endocytosis, caveolar endocytosis, and macropinocytosis are the most researched routes in the field of drug delivery. In addition to conventional small therapeutic molecules, the use of nanoformulations and large molecules, such as nucleic acids, peptides, and antibodies, have broadened the field of drug delivery. Although the majority of small therapeutic molecules can enter cells via passive diffusion, large molecules, and advanced targeted delivery systems, such as nanoparticles, are internalized by the endocytic route. Therefore, it is imperative to understand the characteristics of the endocytic routes in greater detail to design therapeutic molecules or formulations for successful delivery to the intracellular targets. This review highlights the prospects and limitations of the major endocytic routes for drug delivery, with a major emphasis on macropinocytosis. Since macropinocytosis is a non-selective uptake of extracellular matrix, the selective induction of macropinocytosis, using compounds that induce macropinocytosis and modulate macropinosome trafficking pathways, could be a potential approach for the intracellular delivery of diverse therapeutic modalities. Furthermore, we have summarized the characteristics associated with the formulations or drug carriers that can affect the endocytic routes for cellular internalization. The techniques that are used to study the intracellular uptake processes of therapeutic molecules are briefly discussed. Finally, the major limitations for intracellular targeting, endo-lysosomal degradation, and different approaches that have been used in overcoming these limitations, are highlighted in this review.
Collapse
Affiliation(s)
- Rabin Neupane
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Chandrabose Karthikeyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak 484887, India
| | - Charles R Asbhy
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY 10049, USA
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, AL 36849, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
41
|
Li T, Wang Y, Chen X, Cui H, Zhang L, Liu J, Wang J, Wang X, Zhao Y, Chen Q, Wang J. Direct Cytosolic Delivery of siRNA Conjugates: A Paradigm in Antiangiogenic Therapy for Choroidal Neovascularization. ACS NANO 2025; 19:11249-11262. [PMID: 40072892 DOI: 10.1021/acsnano.4c18924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Small interfering RNA (siRNA) has garnered tremendous interest as a potential therapeutic tool because of its intriguing gene-silencing ability. Toward the success in the manufacture of siRNA therapeutics for the potential treatment of choroidal neovascularization (CNV), siRNA conjugated with dual functional units of membrane-penetrating heptafluoropropyl and age-related macular degeneration-targeting cyclic Arg-Gly-Asp (RGD) peptide was attempted for transcellular transportation into the cell interiors. Of note, cyclic RGD allowed selective affinities toward the angiogenic endothelial cells in the pathological CNV. Noteworthy is the functional heptafluoropropyl group, due to its tempting lipophobic and hydrophobic properties, stimulating energy-independent transcellular trafficking behaviors to the cytoplasm directly from the extracellular compartments, namely, the nonendocytotic pathway. The behaviors manage to avoid the well-acknowledged drawback of endolysosomal entrapment, which is deemed to be the critical threat to the biovulnerable genomic therapeutics, thereby contributing to potent gene knockdown at the affected cells. Aiming for treatment of CNV, the siRNA duo was schemed with appropriate chemistry-based modifications for the targeted knockdown of both angiogenic VEGF-A and VEGF-R2. Subsequent investigations verified the potent reduction of vascular leakage, and our proposed siRNA duo accomplished a significant reduction of 67.3% in the mean area of the CNV lesion. Bioinformatic analysis has unveiled a multitude of therapeutic benefits conferred by anti-VEGF therapy, extending beyond the mere inhibition of angiogenesis, including the regulated leukocyte transendothelial migration, retinol metabolism, and estrogen signaling pathways. Hence, our proposed chemistry represents an interesting siRNA conjugate strategy accomplishing direct intracellular transportation of macromolecular biological payloads to the cytosol. Hence, this proposed fluorination strategy should be highlighted to encourage the development of appropriate prodrug chemistry in pursuit of transcellular trafficking of membrane-impermissible and biovulnerable biotherapeutics.
Collapse
Affiliation(s)
- Tongqi Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Yue Wang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
| | - Xiyi Chen
- School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, China
| | - Hongyan Cui
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Liuwei Zhang
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Jun Liu
- School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Jin Wang
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, No. 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Xiumei Wang
- School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
| | - Qixian Chen
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzheng 518120, China
| | - Jing Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| |
Collapse
|
42
|
Won JE, Park M, Hong SH, Kim YS, Song H. Quantum dots as biocompatible small RNA nanocarriers modulating macrophage polarization to treat Asherman's syndrome. NPJ Regen Med 2025; 10:15. [PMID: 40133362 PMCID: PMC11937404 DOI: 10.1038/s41536-025-00403-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Macrophages play a key role in host defense and inflammation, with polarization ranging from pro-inflammatory M1 to anti-inflammatory M2 states. However, effective modulation of macrophage polarity via nucleotide delivery is challenging. This study developed polyethyleneimine-modified carboxyl quantum dots (QDP) as a biocompatible carrier for small RNA delivery to modulate macrophage polarization. QDP-mediated delivery of miR-10a (QDP/miR-10a) rebalanced macrophage polarity and alleviated uterine inflammation and fibrosis in a mouse model of Asherman's syndrome (AS). In vitro, QDP effectively delivered small RNA into RAW 264.7 cells without cytotoxicity, converting LPS-induced M1 to M2 macrophages by inhibiting NF-κB, MAPK, and AKT signaling. In vivo, QDP/miR-10a reduced M1 macrophages, restored polarization, and enhanced uterine restoration in AS mice without affecting systemic immunity. Thus, QDP represents a safe and effective nanocarrier for small RNA delivery to modulate macrophage polarization for inflammatory disease treatment, including AS.
Collapse
Affiliation(s)
- Ji Eun Won
- Department of Life Science, Graduate School, CHA University, Seongnam, 13488, Republic of Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, 13488, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Kangwon, 24341, Republic of Korea
- KW-Bio Co., Ltd, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Yeon Sun Kim
- Department of Biomedical Science, CHA University, Seongnam, 13488, Republic of Korea.
| | - Haengseok Song
- Department of Life Science, Graduate School, CHA University, Seongnam, 13488, Republic of Korea.
- Department of Biomedical Science, CHA University, Seongnam, 13488, Republic of Korea.
- KW-Bio Co., Ltd, Chuncheon, Gangwon, 24252, Republic of Korea.
- Division of Life Science, CHA University, Seongnam, 13488, Republic of Korea.
- CHA Advanced Research Institute, Seongnam, 13488, Republic of Korea.
| |
Collapse
|
43
|
de Oliveira Junior ER, Silva JM, Salomão MA, de Almeida Oliveira NC, de Freitas CS, Ferreira NN, Moreno NS, Rodero CF, Graziani D, Zucolotto V, Mendanha SA, Lima EM. Optimized mucus adhesion and penetration of lipid-polymer nanoparticles enables effective nose-to-brain delivery of perillyl alcohol for glioblastoma therapy. Drug Deliv Transl Res 2025:10.1007/s13346-025-01837-5. [PMID: 40133569 DOI: 10.1007/s13346-025-01837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 03/27/2025]
Abstract
The delivery of drugs directly from the nose to the brain has been explored for the treatment of neurological diseases, such as glioblastoma, by overcoming the blood-brain barrier. Nanocarriers have demonstrated outstanding ability to enhance drug bioavailability in the brain, following intranasal administration. However, the performance of these nanosystems may be hindered by inadequate interactions with the nasal mucosa, limiting their effectiveness in reaching the olfactory region, and consequently, the translocation of particles to the brain. Here, we designed hybrid lipid-polymer nanoparticles (LPNP), containing the cationic lipid DOTAP and the triblock copolymer Pluronic® F127 to combine the mucoadhesiveness and mucus-penetrating properties. Perillyl alcohol (POH), a molecule currently under clinical trials against glioblastoma, via intranasal route, was entrapped in the nanoparticles. LPNP-POH exhibited a balanced profile of mucus adhesion and penetration, suggesting that the formulation may enhance mucosal retention while maintaining effective mucus diffusivity. In vivo evaluations displayed higher translocation of LPNP-POH from the nasal cavity to the brain. LPNP-POH resulted in a 2.5-fold increase in the concentration of perillyl acid (a primary metabolite of POH) in the cerebral tissue compared to the free drug. In vitro assays demonstrated that LPNP-POH increased the cytotoxicity and reduced the tumor growth of U87MG glioma cells. These results highlighted that the engineered formulation, with optimized mucoadhesiveness and mucus penetration properties, improved nose-to-brain delivery of POH, offering a promising potential for glioblastoma therapy.
Collapse
Affiliation(s)
- Edilson Ribeiro de Oliveira Junior
- Farmatec - Laboratory for RD&I in Pharmaceutical Nanotechnology and Drug Delivery Systems, Samambaia Technology Park, UFG, Goiás, Goiânia, 74690-631, Brazil
| | | | - Mariana Arraes Salomão
- Farmatec - Laboratory for RD&I in Pharmaceutical Nanotechnology and Drug Delivery Systems, Samambaia Technology Park, UFG, Goiás, Goiânia, 74690-631, Brazil
- School of Pharmacy, Federal University of Goiás, Goiânia, 74690-631, Brazil
| | - Nathalia Correa de Almeida Oliveira
- Farmatec - Laboratory for RD&I in Pharmaceutical Nanotechnology and Drug Delivery Systems, Samambaia Technology Park, UFG, Goiás, Goiânia, 74690-631, Brazil
- School of Pharmacy, Federal University of Goiás, Goiânia, 74690-631, Brazil
| | - Carla Santos de Freitas
- Farmatec - Laboratory for RD&I in Pharmaceutical Nanotechnology and Drug Delivery Systems, Samambaia Technology Park, UFG, Goiás, Goiânia, 74690-631, Brazil
| | - Natália Noronha Ferreira
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo, 13566-590, Brazil
| | - Natalia Sanchez Moreno
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo, 13566-590, Brazil
| | - Camila Fernanda Rodero
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo, 13566-590, Brazil
| | - Daniel Graziani
- Laboratory of Molecular, Cell and Tissue Analysis, School of Veterinary and Animal Sciences, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Valtencir Zucolotto
- Nanomedicine and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo, 13566-590, Brazil
| | - Sebastião Antônio Mendanha
- Farmatec - Laboratory for RD&I in Pharmaceutical Nanotechnology and Drug Delivery Systems, Samambaia Technology Park, UFG, Goiás, Goiânia, 74690-631, Brazil
- Institute of Physics, Federal University of Goiás, Goiânia, Goiás, 74690-900, Brazil
| | - Eliana Martins Lima
- Farmatec - Laboratory for RD&I in Pharmaceutical Nanotechnology and Drug Delivery Systems, Samambaia Technology Park, UFG, Goiás, Goiânia, 74690-631, Brazil.
- School of Pharmacy, Federal University of Goiás, Goiânia, 74690-631, Brazil.
| |
Collapse
|
44
|
Schorr K, Beck S, Zimmer O, Baumann F, Keller M, Witzgall R, Goepferich A. The quantity of ligand-receptor interactions between nanoparticles and target cells. NANOSCALE HORIZONS 2025; 10:803-823. [PMID: 39951050 DOI: 10.1039/d4nh00645c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Achieving high target cell avidity in combination with cell selectivity are fundamental, but largely unachieved goals in the development of biomedical nanoparticle systems, which are intricately linked to the quantity of targeting functionalities on their surface. Viruses, regarded as almost ideal role models for nanoparticle design, are evolutionary optimized, so that they cope with this challenge bearing an extremely low number of spikes, and thus binding domains, on their surface. In comparison, nanoparticles are usually equipped with more than an order of magnitude more ligands. It is therefore obvious that one key factor for increasing nanoparticle efficiency in terms of avidity and selectivity lies in optimizing their ligand number. A first step along this way is to know how many ligands per nanoparticle are involved in specific binding with target cell receptors. This question is addressed experimentally for a block copolymer nanoparticle model system. The data confirm that only a fraction of the nanoparticle ligands is involved in the binding processes: with a total ligand valency of 29 ligands/100 nm2 surface area a maximum 5.3 ligands/100 nm2 are involved in specific receptor binding. This corresponds to an average number of 251 binding ligands per nanoparticle, a number that can be rationalized within the biological context of the model system.
Collapse
Affiliation(s)
- Kathrin Schorr
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Sebastian Beck
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Oliver Zimmer
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Felix Baumann
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, D-93040 Regensburg, Germany.
| | - Ralph Witzgall
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, 93053 Regensburg, Bavaria, Germany.
| |
Collapse
|
45
|
Stukan I, Żuk A, Pukacka K, Mierzejewska J, Pawłowski J, Kowalski B, Dąbkowska M. Wolf in Sheep's Clothing: Taming Cancer's Resistance with Human Serum Albumin? Int J Nanomedicine 2025; 20:3493-3525. [PMID: 40125439 PMCID: PMC11930253 DOI: 10.2147/ijn.s500997] [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: 11/05/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Human serum albumin (HSA) has emerged as a promising carrier for nanodrug delivery, offering unique structural properties that can be engineered to overcome key challenges in cancer treatment, especially resistance to chemotherapy. This review focuses on the cellular uptake of albumin-based nanoparticles and the modifications that enhance their ability to bypass resistance mechanisms, particularly multidrug resistance type 1 (MDR1), by improving targeting to cancer cells. In our unique approach, we integrate the chemical properties of albumin, its interactions with cancer cells, and surface modifications of albumin-based delivery systems that enable to bypass resistance mechanisms, particularly those related to MDR1, and precisely target receptors on cancer cells to improve treatment efficacy. We discuss that while well-established albumin receptors such as gp60 and gp18/30 are crucial for cellular uptake and transcytosis, their biology remains underexplored, limiting their translational potential. Additionally, we explore the potential of emerging targets, such as cluster of differentiation 44 (CD44), cluster of differentiation (CD36) and transferrin receptor TfR1, as well as the advantages of using dimeric forms of albumin (dHSA) to further enhance delivery to resistant cancer cells. Drawing from clinical examples, including the success of albumin-bound paclitaxel (Abraxane) and new formulations like Pazenir and Fyarro (for Sirolimus), we identify gaps in current knowledge and propose strategies to optimize albumin-based systems. In conclusion, albumin-based nanoparticles, when tailored with appropriate modifications, have the potential to bypass multidrug resistance and improve the targeting of cancer cells. By enhancing albumin's ability to efficiently deliver therapeutic agents, these carriers represent a promising approach to addressing one of oncology's most persistent challenges, with substantial potential to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Iga Stukan
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Anna Żuk
- Independent Laboratory of Community Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Kamila Pukacka
- Department of Pharmaceutical Technology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Julia Mierzejewska
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Jakub Pawłowski
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Bogusław Kowalski
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Maria Dąbkowska
- Independent Laboratory of Pharmacokinetic and Clinical Pharmacy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
46
|
Kumar A, Shahvej SK, Yadav P, Modi U, Yadav AK, Solanki R, Bhatia D. Clinical Applications of Targeted Nanomaterials. Pharmaceutics 2025; 17:379. [PMID: 40143042 PMCID: PMC11944548 DOI: 10.3390/pharmaceutics17030379] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Targeted nanomaterials are at the forefront of advancements in nanomedicine due to their unique and versatile properties. These include nanoscale size, shape, surface chemistry, mechanical flexibility, fluorescence, optical behavior, magnetic and electronic characteristics, as well as biocompatibility and biodegradability. These attributes enable their application across diverse fields, including drug delivery. This review explores the fundamental characteristics of nanomaterials and emphasizes their importance in clinical applications. It further delves into methodologies for nanoparticle programming alongside discussions on clinical trials and case studies. We discussed some of the promising nanomaterials, such as polymeric nanoparticles, carbon-based nanoparticles, and metallic nanoparticles, and their role in biomedical applications. This review underscores significant advancements in translating nanomaterials into clinical applications and highlights the potential of these innovative approaches in revolutionizing the medical field.
Collapse
Affiliation(s)
- Ankesh Kumar
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - SK Shahvej
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Pankaj Yadav
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Unnati Modi
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Amit K. Yadav
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
47
|
Larreina Vicente N, Srinivas M, Tagit O. Perfluorocarbon-Loaded Poly(lactide- co-glycolide) Nanoparticles from Core to Crust: Multifaceted Impact of Surfactant on Particle Ultrastructure, Stiffness, and Cell Uptake. ACS APPLIED POLYMER MATERIALS 2025; 7:2864-2878. [PMID: 40110246 PMCID: PMC11915196 DOI: 10.1021/acsapm.4c03360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Poly(lactide-co-glycolide) nanoparticles (PLGA NPs) loaded with Perfluoro-15-crown-5-ether (PFCE) have been developed for imaging applications. A slight modification of the formulation led to the formation of two distinct particle ultrastructures: multicore particles (MCPs) and core-shell particles (CSPs), where poly(vinyl alcohol) (PVA), a nonionic surfactant, and sodium cholate (NaCh), an anionic surfactant, were used, respectively. Despite their similar composition and colloidal characteristics, these particles have previously demonstrated significant differences in their in vivo distribution and clearance. We hypothesize that these differences are collectively driven by variations in their structural, chemical, and mechanical properties, which are investigated in this study. Nanomechanical characterizations of MCPs and CSPs by atomic force microscopy (AFM) revealed elastic modulus values of 54 and 270 MPa in water, respectively, indicating a better permeability and deformability of the multicore ultrastructure. The impact of the surfactant on the NP surface chemistry was evidenced by their protein corona, which was significantly greater in the CSPs. Additionally, an important amount of residual NaCh was found on the surface of CSPs, which formed strong interactions with bovine serum albumin (BSA), accounting for the difference in protein coronas and surface chemistry. Surprisingly, in vitro cell uptake studies showed a higher uptake of MCPs by RAW macrophages but a preference for CSPs by HeLa cells. We conclude that for this specific formulation and in this stiffness range, mechanical differences have a stronger impact in HeLa cells, while surface properties and chemical recognition play a more important role in uptake by macrophages. Overall, the extent to which a physical factor impacts cell uptake is highly dependent on the specific uptake mechanism. With this study, we provide an integrated perspective on the role of different surfactants in the particle formation process, their impact on particle ultrastructure, mechanical properties, and surface chemistry, and the overall effect on cell uptake in vitro.
Collapse
Affiliation(s)
- Naiara Larreina Vicente
- Cell Biology and Immunology (CBI), Wageningen University, De Elst 1, Wageningen 6708 WD, Netherlands
| | - Mangala Srinivas
- Cell Biology and Immunology (CBI), Wageningen University, De Elst 1, Wageningen 6708 WD, Netherlands
| | - Oya Tagit
- Group of Biointerfaces, Institute for Chemistry and Bioanalytics, FHNW University of Applied Sciences and Arts Northwestern Switzerland, Hofackerstrasse 30, Muttenz 4132, Switzerland
| |
Collapse
|
48
|
Yadav A, Sharma A, Moulick M, Ghatak S. Nanomanaging Chronic Wounds with Targeted Exosome Therapeutics. Pharmaceutics 2025; 17:366. [PMID: 40143030 PMCID: PMC11945274 DOI: 10.3390/pharmaceutics17030366] [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: 02/12/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic wounds pose a significant healthcare challenge, impacting millions of patients worldwide and burdening healthcare systems substantially. These wounds often occur as comorbidities and are prone to infections. Such infections hinder the healing process, complicating clinical management and proving recalcitrant to therapy. The environment within the wound itself poses challenges such as lack of oxygen, restricted blood flow, oxidative stress, ongoing inflammation, and bacterial presence. Traditional systemic treatment for such chronic peripheral wounds may not be effective due to inadequate blood supply, resulting in unintended side effects. Furthermore, topical applications are often impervious to persistent biofilm infections. A growing clinical concern is the lack of effective therapeutic modalities for treating chronic wounds. Additionally, the chemically harsh wound microenvironment can reduce the effectiveness of treatments, highlighting the need for drug delivery systems that can deliver therapies precisely where needed with optimal dosages. Compared to cell-based therapies, exosome-based therapies offer distinct advantages as a cell-free approach for chronic wound treatment. Exosomes are of endosomal origin and enable cell-to-cell communications, and they possess benefits, including biocompatibility and decreased immunogenicity, making them ideal vehicles for efficient targeting and minimizing off-target damage. However, exosomes are rapidly cleared from the body, making it difficult to maintain optimal therapeutic concentrations at wound sites. The hydrogel-based approach and development of biocompatible scaffolds for exosome-based therapies can be beneficial for sustained release and prolong the presence of these therapeutic exosomes at chronic wound sites. Engineered exosomes have been shown to possess stability and effectiveness in promoting wound healing compared to their unmodified counterparts. Significant progress has been made in this field, but further research is essential to unlock their clinical potential. This review seeks to explore the benefits and opportunities of exosome-based therapies in chronic wounds, ensuring sustained efficacy and precise delivery despite the obstacles posed by the wound environment.
Collapse
Affiliation(s)
| | | | | | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.Y.); (A.S.); (M.M.)
| |
Collapse
|
49
|
Akhtar S, Zuhair F. Advancing Nanomedicine Through Electron Microscopy: Insights Into Nanoparticle Cellular Interactions and Biomedical Applications. Int J Nanomedicine 2025; 20:2847-2878. [PMID: 40078651 PMCID: PMC11899938 DOI: 10.2147/ijn.s500978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Nanomedicine has revolutionized cancer treatment by the development of nanoparticles (NPs) that offer targeted therapeutic delivery and reduced side effects. NPs research in nanomedicine significantly focuses on understanding their cellular interactions and intracellular mechanisms. A precise understanding of nanoparticle interactions at the subcellular level is crucial for their effective application in cancer therapy. Electron microscopy has proven essential, offering high-resolution insights into nanoparticle behavior within biological systems. This article reviews the role of electron microscopy in elucidating the cellular uptake and intracellular interactions of NPs. Transmission electron microscopy (TEM) provides imaging capabilities, such as cryo three-dimensional tomography, which offer in-depth insights into nanoparticle localization, endocytosis pathways, and subcellular interactions, while high resolution-TEM is primarily used for studying the atomic structure of isolated NPs rather than nanoparticles within cells or tissues. On the other hand, scanning electron microscopy (SEM) is ideal for examining larger surface areas and provides a broader perspective on the morphology and topography of the samples. The review highlights the advantages of electron microscopy in visualizing nanoparticle interactions with cellular structures and tracking their mechanisms of action. It also addresses the challenges associated with electron microscopy characterization, such as tedious sample preparation, static imaging limitations, and a restricted field of view. By examining various nanoparticle uptake pathways, and cellular destination of NPs with examples, the article emphasizes the importance of these pathways to optimize nanoparticle design and enhance therapeutic efficacy. This review underscores the need for continued advancement in electron microscopy techniques to improve the effectiveness of nanomedicine and address existing challenges. In summary, electron microscopy is a key tool for advancing our understanding of nanoparticle behavior in biological contexts, aiding in the design and optimization of nanomedicines by providing insights into nanoparticle cellular dynamics, uptake mechanisms, and therapeutic applications.
Collapse
Affiliation(s)
- Sultan Akhtar
- Department of Biophysics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Fatimah Zuhair
- Department of Infection Control, Alzahra General Hospital, Qatif, 31911, Saudi Arabia
| |
Collapse
|
50
|
Kurniawan A, Mahendra I, Febrian MB, Utama MS, Gunadi JW, Wahyudianingsih R, Lesmana R, Halimah I, Sriyani ME, Widyasari EM, Wibawa THA, Rizaludin A, Kusumaningrum CE, Syarif DG. Biological evaluation of hydroxyapatite zirconium nanoparticle as a potential radiosensitizer for lung cancer X-ray induced photodynamic therapy. Appl Radiat Isot 2025; 217:111615. [PMID: 39632318 DOI: 10.1016/j.apradiso.2024.111615] [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: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Photodynamic therapy has been recognized as a viable approach for lung cancer treatment. Some photosensitizer agents are known as X-ray sensitive and could improve radiotherapy efficacy. The use of nanoparticles for drug delivery and as photosensitizer agents offers various advantages because of their rapid cellular accumulation and distribution into target organs. On the other hand, several nanoparticles could trigger adverse effects during cancer treatment. In this article, the biological study of hydroxyapatite zirconium nanoparticles (HApZr) as photosensitizer candidates for X-ray-induced photodynamic therapy has been demonstrated in vitro and in vivo. This nanoparticle increased the intracellular reactive oxygen species (ROS) levels after the delivery of ionizing radiation at 5 Gy to a cancer cell line and showed higher cytotoxicity compared to non-irradiated treatment. In vitro cellular uptake based on cell imaging also indicated a promising intake and an ability to kill cancer cells. Subsequently, an in vivo evaluation using orthotopic lung cancer mouse models also showed their good accumulation in target organs, with lower accumulation in normal lung tissue. Moreover, studies of acute toxicity showed that a dose of 50 μg/mL yielded minor pathological changes on histological evaluations, which were supported by a biochemical analysis. In addition, HApZr nanoparticles also increase TNF-α which enhancing the cytotoxic effect after irradiation. Finally, these findings were important for further investigation of the clinical application of these HApZr nanoparticles for the treatment of patients with lung cancer.
Collapse
Affiliation(s)
- Ahmad Kurniawan
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia.
| | - Isa Mahendra
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Muhamad Basit Febrian
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Marhendra Satria Utama
- Department of Radiology, Faculty of Medicine, Universitas Padjadjaran/Dr. Hasan Sadikin General Hospital, Bandung, Indonesia
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia
| | - Roro Wahyudianingsih
- Department of Pathological Anatomy, Faculty of Medicine, Maranatha Christian University, Bandung, Indonesia
| | - Ronny Lesmana
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Iim Halimah
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Maula Eka Sriyani
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Eva Maria Widyasari
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Teguh Hafiz Ambar Wibawa
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Asep Rizaludin
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Crhisterra Ellen Kusumaningrum
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Dani Gustaman Syarif
- Research Center for Radiation Process Technology, National Research and Innovation Agency, Serpong, Indonesia
| |
Collapse
|