Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Nanobiotechnology ; 22(1): 184, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622644

ABSTRACT

Despite the advent of numerous targeted therapies in clinical practice, anthracyclines, including doxorubicin (DOX), continue to play a pivotal role in breast cancer (BC) treatment. DOX directly disrupts DNA replication, demonstrating remarkable efficacy against BC cells. However, its non-specificity toward cancer cells leads to significant side effects, limiting its clinical utility. Interestingly, DOX can also enhance the antitumor immune response by promoting immunogenic cell death in BC cells, thereby facilitating the presentation of tumor antigens to the adaptive immune system. However, the generation of an adaptive immune response involves highly proliferative processes, which may be adversely affected by DOX-induced cytotoxicity. Therefore, understanding the impact of DOX on dividing T cells becomes crucial, to deepen our understanding and potentially devise strategies to shield anti-tumor immunity from DOX-induced toxicity. Our investigation focused on studying DOX uptake and its effects on human lymphocytes. We collected lymphocytes from healthy donors and BC patients undergoing neoadjuvant chemotherapy (NAC). Notably, patient-derived peripheral blood mononuclear cells (PBMC) promptly internalized DOX when incubated in vitro or isolated immediately after NAC. These DOX-treated PBMCs exhibited significant proliferative impairment compared to untreated cells or those isolated before treatment initiation. Intriguingly, among diverse lymphocyte sub-populations, CD8 + T cells exhibited the highest uptake of DOX. To address this concern, we explored a novel DOX formulation encapsulated in ferritin nanocages (FerOX). FerOX specifically targets tumors and effectively eradicates BC both in vitro and in vivo. Remarkably, only T cells treated with FerOX exhibited reduced DOX internalization, potentially minimizing cytotoxic effects on adaptive immunity.Our findings underscore the importance of optimizing DOX delivery to enhance its antitumor efficacy while minimizing adverse effects, highlighting the pivotal role played by FerOX in mitigating DOX-induced toxicity towards T-cells, thereby positioning it as a promising DOX formulation. This study contributes valuable insights to modern cancer therapy and immunomodulation.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Leukocytes, Mononuclear , Neoadjuvant Therapy , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Antineoplastic Agents/pharmacology , Cell Line, Tumor
2.
Small ; 18(39): e2202145, 2022 09.
Article in English | MEDLINE | ID: mdl-36026572

ABSTRACT

Enzymes are an important component for bottom-up building of synthetic/artificial cells. Nanozymes are nanomaterials with intrinsic enzyme-like properties, however, the construction of synthetic cells using nanozymes is difficult owing to their high surface energy or large size. Herein, the authors show a protein-based general platform that biomimetically integrates various ultrasmall metal nanozymes into protein shells. Specifically, eight metal-based ultrasmall nano-particles/clusters are in situ incorporated into ferritin nanocages that are self-assembled by 24 subunits of ferritin heavy chain. As a nanozyme generator, such a platform is suitable for screening the desired enzyme-like activities, including peroxidase (POD), oxidase (OXD), catalase (CAT) and superoxide dismutase (SOD). After screening, it is found that Ru intrinsically possesses the highest POD-like and CAT-like activities, while Mn and Pt show the highest OXD-like and SOD-like activities, respectively. Additionally, the inducers/inhibitors of various nanozymes are screened from more than 50 compounds to improve or inhibit their enzyme-like activities. Based on the screened nanozymes and their inhibitors, a proof-of-conceptually constructs cell-mimicking catalytic vesicles to mimic or modulate the events of redox homeostasis in living cells. This study offers a type of artificial metalloenzyme based on nanotechnology and shows a choice for bottom-up enzyme-based synthetic cell systems in a fully synthetic manner.


Subject(s)
Apoferritins , Nanostructures , Catalase , Catalysis , Ferritins , Peroxidase , Peroxidases , Superoxide Dismutase
3.
Small ; 13(37)2017 10.
Article in English | MEDLINE | ID: mdl-28786527

ABSTRACT

Ferritins are ubiquitous iron storage proteins where Fe(II) sequestration prevents not only its spontaneous oxidation to Fe(III) but also production of toxic free radicals. Recently, scientists have subverted these nature functions and used ferritin cage structures of nanometer dimensions for encapsulation of guest molecules such as anti-cancer drugs or bioactive nutrients based on pH induced ferritin disassembly and reassembly property. However, prior to this study, ferritin nanocage was required to disassemble only under harsh pH conditions (≤2.0 or ≥11.0), followed by reassembly at near neutral pH. Such harsh conditions can cause protein or guest molecules damage to a great extent during this pH-induced unfolding-refolding process. Here, we provide evidence demonstrating that the apoferritin shell is flexible rather than rigid. Indeed, we found that two large complex molecules, uranyl acetate dihydrate and phosphotungstic acid, can reach the cavity of both plant and animal apoferritin followed by mineralization. Moreover, large organic compound such as curcumin and doxorubicin can also be encapsulated within ferritin cavity by its mixing with protein. This strategy will increase the use of ferritin in nanotechnology, and could be also applicable to other shell-like proteins as templates to prepare nanomaterials.


Subject(s)
Ferritins/chemistry , Nanostructures/chemistry , Particle Size , Apoferritins/chemistry , Curcumin/chemistry , Gallic Acid/chemistry , Nanocomposites/chemistry , Nanocomposites/ultrastructure , Nanostructures/ultrastructure , Scattering, Radiation , Spectrometry, X-Ray Emission
4.
Int J Nanomedicine ; 19: 4263-4278, 2024.
Article in English | MEDLINE | ID: mdl-38766663

ABSTRACT

Introduction: Photodynamic Therapy (PDT) is a promising, minimally invasive treatment for cancer with high immunostimulatory potential, no reported drug resistance, and reduced side effects. Indocyanine Green (ICG) has been used as a photosensitizer (PS) for PDT, although its poor stability and low tumor-target specificity strongly limit its efficacy. To overcome these limitations, ICG can be formulated as a tumor-targeting nanoparticle (NP). Methods: We nanoformulated ICG into recombinant heavy-ferritin nanocages (HFn-ICG). HFn has a specific interaction with transferrin receptor 1 (TfR1), which is overexpressed in most tumors, thus increasing HFn tumor tropism. First, we tested the properties of HFn-ICG as a PS upon irradiation with a continuous-wave diode laser. Then, we evaluated PDT efficacy in two breast cancer (BC) cell lines with different TfR1 expression levels. Finally, we measured the levels of intracellular endogenous heavy ferritin (H-Fn) after PDT treatment. In fact, it is known that cells undergoing ROS-induced autophagy, as in PDT, tend to increase their ferritin levels as a defence mechanism. By measuring intracellular H-Fn, we verified whether this interplay between internalized HFn and endogenous H-Fn could be used to maximize HFn uptake and PDT efficacy. Results: We previously demonstrated that HFn-ICG stabilized ICG molecules and increased their delivery to the target site in vitro and in vivo for fluorescence guided surgery. Here, with the aim of using HFn-ICG for PDT, we showed that HFn-ICG improved treatment efficacy in BC cells, depending on their TfR1 expression. Our data revealed that endogenous H-Fn levels were increased after PDT treatment, suggesting that this defence reaction against oxidative stress could be used to enhance HFn-ICG uptake in cells, increasing treatment efficacy. Conclusion: The strong PDT efficacy and peculiar Trojan horse-like mechanism, that we revealed for the first time in literature, confirmed the promising application of HFn-ICG in PDT.


Subject(s)
Breast Neoplasms , Indocyanine Green , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Female , Humans , Antigens, CD/metabolism , Apoferritins/chemistry , Breast Neoplasms/therapy , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Ferritins/chemistry , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Indocyanine Green/therapeutic use , MCF-7 Cells , Nanoparticles/chemistry , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Receptors, Transferrin/metabolism
5.
Pharmaceutics ; 16(3)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38543296

ABSTRACT

Chronic lymphocytic leukemia (CLL) is a widespread type of leukemia that predominantly targets B lymphocytes, undermining the balance between cell proliferation and apoptosis. In healthy B cells, miR-15/16, a tandem of microRNAs, functions as a tumor suppressor, curbing the expression of the antiapoptotic B cell lymphoma 2 protein (Bcl-2). Conversely, in CLL patients, a recurring deletion on chromosome 13q14, home to the miR15-a and miR16-1 genes, results in Bcl-2 overexpression, thereby fostering the onset of the pathology. In the present research, a novel approach utilizing humanized ferritin-based nanoparticles was employed to successfully deliver miR15-a and miR-16-1 into MEG01 cells, a model characterized by the classic CLL deletion and overexpression of the human ferritin receptor (TfR1). The loaded miR15-a and miR16-1, housed within modified HumAfFt, were efficiently internalized via the MEG01 cells and properly directed into the cytoplasm. Impressively, the concurrent application of miR15-a and miR16-1 demonstrated a robust capacity to induce apoptosis through the reduction in Bcl-2 expression levels. This technology, employing RNA-loaded ferritin nanoparticles, hints at promising directions in the battle against CLL, bridging the substantial gap left by traditional transfection agents and indicating a pathway that may offer hope for more effective treatments.

6.
Int J Biol Macromol ; 277(Pt 2): 134373, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094874

ABSTRACT

Ferritins are natural proteins which spontaneously self-assemble forming hollow nanocages physiologically deputed to iron storage and homeostasis. Thanks to their high stability and easy production in vitro, ferritins represent an intriguing system for nanobiotechnology. Here we investigated the mechanism of disassembly and reassembly of a human recombinant ferritin constituted by the heavy chain (hHFt) exploiting a new procedure which involves the use of minimal amounts of sodium dodecyl sulfate (SDS) and assessed its effectiveness in comparison with two commonly used protocols based on pH shift at highly acidic and alkaline values. The interest in this ferritin as drug nanocarrier is related to the strong affinity of the human H-chain for the transferrin receptor TfR-1, overexpressed in several tumoral cell lines. Using different techniques, like NMR, TEM and DLS, we demonstrated that the small concentrations of SDS can eliminate the nanocage architecture without detaching the monomers from each other, which instead remain strongly associated. Following this procedure, we encapsulated into the nanocage a small ruthenium complex with a remarkable improvement with respect to previous protocols in terms of yield, structural integrity of the recovered protein and encapsulation efficiency. In our opinion, the extensive network of interchain interactions preserved during the SDS-based disassembly procedure represents the key for a complete and correct hHFt reassembly.


Subject(s)
Drug Carriers , Ferritins , Humans , Ferritins/chemistry , Drug Carriers/chemistry , Receptors, Transferrin/metabolism , Receptors, Transferrin/chemistry , Nanoparticles/chemistry , Drug Delivery Systems , Hydrogen-Ion Concentration , Recombinant Proteins/chemistry , Sodium Dodecyl Sulfate/chemistry , Antigens, CD
7.
Sci Rep ; 14(1): 11533, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773170

ABSTRACT

Tauopathies, including Alzheimer's disease and Frontotemporal Dementia, are debilitating neurodegenerative disorders marked by cognitive decline. Despite extensive research, achieving effective treatments and significant symptom management remains challenging. Accurate diagnosis is crucial for developing effective therapeutic strategies, with hyperphosphorylated protein units and tau oligomers serving as reliable biomarkers for these conditions. This study introduces a novel approach using nanotechnology to enhance the diagnostic process for tauopathies. We developed humanized ferritin nanocages, a novel nanoscale delivery system, designed to encapsulate and transport a tau-specific fluorophore, BT1, into human retinal cells for detecting neurofibrillary tangles in retinal tissue, a key marker of tauopathies. The delivery of BT1 into living cells was successfully achieved through these nanocages, demonstrating efficient encapsulation and delivery into retinal cells derived from human induced pluripotent stem cells. Our experiments confirmed the colocalization of BT1 with pathological forms of tau in living retinal cells, highlighting the method's potential in identifying tauopathies. Using ferritin nanocages for BT1 delivery represents a significant contribution to nanobiotechnology, particularly in neurodegenerative disease diagnostics. This method offers a promising tool for the early detection of tau tangles in retinal tissue, with significant implications for improving the diagnosis and management of tauopathies. This study exemplifies the integration of nanotechnology with biomedical science, expanding the frontiers of nanomedicine and diagnostic techniques.


Subject(s)
Ferritins , Retina , Tauopathies , tau Proteins , Humans , tau Proteins/metabolism , Ferritins/metabolism , Retina/metabolism , Retina/pathology , Tauopathies/metabolism , Tauopathies/pathology , Tauopathies/diagnosis , Induced Pluripotent Stem Cells/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology
8.
Food Chem ; 411: 135437, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-36701920

ABSTRACT

Ferritin proteins are promising nano-carriers for bioactive compound delivery. However, the disassembly properties under acidic/alkaline conditions pose risks of cargo leakage. Herein, genipin-mediated chemical crosslinking method was provided as an alternative and effective strategy to construct robust ferritin nanocarrier through controlled-intramolecular conjugation. As indicated by SDS-/Native- PAGE, the crosslinking degree gradually increased with incubating time prolonging. CD results showed that the cross-linking would decrease α-helix content from 78.4 % to 52.7 % upon 6 h incubation. However, TEM images showed that the genipin-modification has subtle influence on its shell-like structure. Remarkably, the cross-linking can be well controlled by intramolecular subunit-subunit conjugation rather than intermolecular conjugation, giving an excellent monodispersity. Importantly, the covalent cross-linking can tight neighboring subunits and inhibit its disassociation, finally inhibiting the leakage of encapsulated-cargos from ferritin cavity under acidic environments. Such findings suggested that the genipin-mediated cross-linking strategy can fabricate robust nano-carriers for bioactive compound delivery.


Subject(s)
Ferritins , Iridoids , Cross-Linking Reagents/chemistry , Iridoids/chemistry , Electrophoresis, Polyacrylamide Gel
9.
Int J Biol Macromol ; 253(Pt 3): 126973, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37729988

ABSTRACT

Ferritin possesses a stable and uniform cage structure, along with tumor-targeting properties and excellent biocompatibility, making it a promising drug delivery vehicle. However, the current ferritin drug loading strategy involves complex steps and harsh reaction conditions, resulting in low yield and recovery of drug loading, which limits the clinical application prospects of ferritin nanomedicine. In this study, we utilized the high-efficiency heat-sensitivity of the multiple channel switch structures of the E-helix-cut ferritin mutant (Ecut-HFn) and Cu2+ assistance to achieve high-efficiency loading of chemotherapeutic drugs in a one-step process at low temperatures. This method features mild reaction conditions (45 °C), high loading efficiency (about 110 doxorubicin (Dox) per Ecut-HFn), and improved protein and Dox recovery rates (with protein recovery rate around 94 % and Dox recovery rate reaching up to 45 %). The prepared ferritin-Dox particles (Ecut-HFn-Cu-Dox) exhibit a uniform size distribution, good stability, and retain the natural tumor targeting ability of ferritin. Overall, this temperature-controlled drug loading strategy utilizing heat-sensitivity ferritin mutants is energy-saving, environmentally friendly, efficient, and easy to operate, offering a new perspective for scaling up the industrial production of ferritin drug carriers.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Humans , Antineoplastic Agents/chemistry , Ferritins/genetics , Ferritins/chemistry , Hot Temperature , Doxorubicin/chemistry , Neoplasms/drug therapy , Drug Carriers/chemistry , Drug Delivery Systems , Nanoparticles/chemistry
10.
Front Mol Biosci ; 10: 1008985, 2023.
Article in English | MEDLINE | ID: mdl-36714262

ABSTRACT

For their easy and high-yield recombinant production, their high stability in a wide range of physico-chemical conditions and their characteristic hollow structure, ferritins (Fts) are considered useful scaffolds to encapsulate bioactive molecules. Notably, for the absence of immunogenicity and the selective interaction with tumor cells, the nanocages constituted by the heavy chain of the human variant of ferritin (hHFt) are optimal candidates for the delivery of anti-cancer drugs. hHFt nanocages can be disassembled and reassembled in vitro to allow the loading of cargo molecules, however the currently available protocols present some relevant drawbacks. Indeed, protein disassembly is achieved by exposure to extreme pH (either acidic or alkaline), followed by incubation at neutral pH to allow reassembly, but the final protein recovery and homogeneity are not satisfactory. Moreover, the exposure to extreme pH may affect the structure of the molecule to be loaded. In this paper, we report an alternative, efficient and reproducible procedure to reversibly disassemble hHFt under mild pH conditions. We demonstrate that a small amount of sodium dodecyl sulfate (SDS) is sufficient to disassemble the nanocage, which quantitatively reassembles upon SDS removal. Electron microscopy and X-ray crystallography show that the reassembled protein is identical to the untreated one. The newly developed procedure was used to encapsulate two small molecules. When compared to the existing disassembly/reassembly procedures, our approach can be applied in a wide range of pH values and temperatures, is compatible with a larger number of cargos and allows a higher protein recovery.

11.
Biotechnol Adv ; 61: 108037, 2022 12.
Article in English | MEDLINE | ID: mdl-36152892

ABSTRACT

Bioactive or nutraceutical ingredients have been widely used in pursuit of health and well-being. However, the environmental instability, poor solubility and bioavailability, and unspecific delivery highly limited their practical values. By virtue of the unique shell-like structure, definite disassembly/reassembly behavior, and excellent safety profile of ferritin protein, it stands out among of various nano-materials and is emerging as one of the most promising vehicles for the encapsulation and delivery of bioactive ingredients or drugs. In this review, we present a systematic overview of recent advances of ferritin-based delivery systems from single-encapsulation, co-encapsulation, to compartmentalized-encapsulation of bioactive ingredients or drugs. Different encapsulation strategies for cargo loading as well as their advantages and drawbacks have been critically reviewed. This study emphasized the importance of the construction of compartmentalized delivery systems through the usage of ferritin nanocages, which exhibit great potential for facilitating the synergistic functionality of different types of cargos. Lastly, the applications of ferritin nanocages for physicochemical improvements and functionality achievements of loaded cargos are summarized. In conclusion, ferritin protein nanocages not only are excellent nanocarriers, but also can act as"multi-seated" vehicles for co-encapsulation and compartmentalized encapsulation of different cargos simultaneously.


Subject(s)
Dietary Supplements , Ferritins , Biological Availability , Excipients , Ferritins/chemistry , Ferritins/metabolism , Solubility , Systematic Reviews as Topic
12.
J Control Release ; 279: 326-335, 2018 06 10.
Article in English | MEDLINE | ID: mdl-29679665

ABSTRACT

Exosomes are small membrane vesicles secreted by most cell types that play an important role in intercellular communication. Due to the characteristic of transferring their biomacromolecules, exosomes have potential as a new alternative for delivering protein therapeutics. Here, we investigate whether exosomes provide crucial advantages over other nanoparticles, in particular protein nanocage formulations, as a delivery system for membrane protein therapeutics. We characterized membrane-scaffold-based exosomes and protein-scaffold-based ferritin nanocages, both harboring SIRPα (signal regulatory protein α), an antagonist of CD47 on tumor cells. The efficacy of these two systems in delivering protein therapeutics was compared by testing their ability to enhance phagocytosis of tumor cells by bone-marrow-derived macrophages and subsequent inhibition of in vivo tumor growth. These analyses allowed us to comprehensively conclude that the therapeutic index of exosome-mediated CD47 blockade against tumor growth inhibition was higher than that of the same dose of ferritin-SIRPα. The results of this analysis reveal the importance of the unique characteristics of exosomes, in particular their membrane scaffold, in improving therapeutic protein delivery compared with protein-scaffold-based nanocages.


Subject(s)
Antigens, Differentiation/administration & dosage , CD47 Antigen/antagonists & inhibitors , Drug Delivery Systems , Nanoparticles , Receptors, Immunologic/administration & dosage , Animals , Antigens, Differentiation/metabolism , Colonic Neoplasms/drug therapy , Exosomes/chemistry , Ferritins/chemistry , HT29 Cells , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phagocytosis , Receptors, Immunologic/metabolism , Xenograft Model Antitumor Assays
13.
J Control Release ; 267: 172-180, 2017 Dec 10.
Article in English | MEDLINE | ID: mdl-28821462

ABSTRACT

Ferritin nanocages are of particular interest as a novel platform for drug and vaccine delivery, diagnosis, biomineralization scaffold and more, due to their perfect and complex symmetry, ideal physical properties, high biocompatibility, low toxicity profiles as well as easy manipulation by genetic or chemical strategies. However, a short half-life is still a hurdle for the translation of ferritin-based nanomedicines into the clinic. Here, we developed a series of rationally designed long circulating ferritin nanocages (LCFNs) with 'Intrinsically Disordered Proteins (IDP)' as a stealth layer for extending the half-life of ferritin nanocages. Through predictions with 3D modelling, the LCFNs were designed, generated and their pharmacokinetic parameters including half-life, clearance rate, mean residence time, and more, were evaluated by qualitative and quantitative analysis. LCFNs have a tenfold increased half-life and overall improved pharmacokinetic parameters compared to wild-type ferritin nanocages (wtFN), corresponding to the low binding against bone marrow-derived macrophages (BMDMs) and endothelial cells. Subsequently, a tumor targeting moiety, epidermal growth factor receptor (EGFR)-targeting affibody peptide, was fused to LCFNs for evaluating their potential as a theragnostic platform. The tumor targeting-LCFNs successfully accumulated to the tumor tissue, by efficient targeting via active and passive properties, and also the shielding effect of IDP in vivo. This strategy can be applied to other protein-based nanocages for further progressing their use in the field of nanomedicine.


Subject(s)
Drug Delivery Systems , Ferritins/administration & dosage , Intrinsically Disordered Proteins/administration & dosage , Nanostructures/administration & dosage , Neoplasms/metabolism , Peptides/administration & dosage , Animals , Ferritins/chemistry , Ferritins/pharmacokinetics , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/pharmacokinetics , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Nanostructures/chemistry , Nanostructures/ultrastructure , Peptides/chemistry , Peptides/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL