Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.426
Filtrar
1.
Food Res Int ; 186: 114344, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729696

RESUMEN

The research aimed to evaluate the effect of ultrasonication and succinylation on the functional, iron binding, physiochemical, and cellular mineral uptake efficacy of chickpea protein concentrate. Succinylation resulted in significant improvements in the water-holding capacity (WHC) (25.47 %), oil-holding capacity (OHC) (31.38 %), and solubility (5.80 %) of the chickpea protein-iron complex. Mineral bioavailability significantly increased by 4.41 %, and there was a significant increase in cellular mineral uptake (64.64 %), retention (36.68 %), and transport (27.96 %). The ferritin content of the succinylated chickpea protein-iron complex showed a substantial increase of 66.31%. Furthermore, the dual modification approach combining ultrasonication and succinylation reduced the particle size of the protein-iron complex with a substantial reduction of 83.25 %. It also resulted in a significant enhancement of 51.5 % in the SH (sulfhydryl) content and 48.92 % in the surface hydrophobicity. Mineral bioavailability and cellular mineral uptake, retention, and transport were further enhanced through dual modification. In terms of application, the addition of single and dual-modified chickpea protein-iron complex to a fruit-based smoothie demonstrated positive acceptance in sensory attributes. Overall, the combined approach of succinylation and ultrasonication to the chickpea protein-iron complex shows a promising strategy for enhancing the physiochemical and techno-functional characteristics, cellular mineral uptake, and the development of vegan food products.


Asunto(s)
Disponibilidad Biológica , Cicer , Hierro , Cicer/química , Hierro/química , Hierro/metabolismo , Humanos , Alimentos Fortificados , Proteínas de Plantas/química , Digestión , Minerales/química , Células CACO-2 , Ácido Succínico/química , Tamaño de la Partícula , Manipulación de Alimentos/métodos , Solubilidad , Ferritinas/química , Ferritinas/metabolismo
2.
Nat Commun ; 15(1): 3802, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714719

RESUMEN

The interaction between nuclear receptor coactivator 4 (NCOA4) and the iron storage protein ferritin is a crucial component of cellular iron homeostasis. The binding of NCOA4 to the FTH1 subunits of ferritin initiates ferritinophagy-a ferritin-specific autophagic pathway leading to the release of the iron stored inside ferritin. The dysregulation of NCOA4 is associated with several diseases, including neurodegenerative disorders and cancer, highlighting the NCOA4-ferritin interface as a prime target for drug development. Here, we present the cryo-EM structure of the NCOA4-FTH1 interface, resolving 16 amino acids of NCOA4 that are crucial for the interaction. The characterization of mutants, designed to modulate the NCOA4-FTH1 interaction, is used to validate the significance of the different features of the binding site. Our results explain the role of the large solvent-exposed hydrophobic patch found on the surface of FTH1 and pave the way for the rational development of ferritinophagy modulators.


Asunto(s)
Microscopía por Crioelectrón , Ferritinas , Coactivadores de Receptor Nuclear , Ferritinas/metabolismo , Ferritinas/química , Ferritinas/genética , Humanos , Coactivadores de Receptor Nuclear/metabolismo , Coactivadores de Receptor Nuclear/química , Coactivadores de Receptor Nuclear/genética , Unión Proteica , Sitios de Unión , Hierro/metabolismo , Autofagia , Modelos Moleculares , Células HEK293 , Oxidorreductasas/metabolismo , Oxidorreductasas/química , Oxidorreductasas/genética , Proteolisis , Mutación
3.
Int J Biol Macromol ; 267(Pt 1): 131492, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38604418

RESUMEN

Human heavy chain ferritin (HFn) protein cage has been explored as a nanocarrier for targeted anticancer drug delivery. Here, we introduced a matrix metalloproteinases (MMPs)-cleavable sequence into the DE loop of HFn, creating an MMP-responsive variant, MR-HFn, for localized and extracellular drug release. The crystal structure of MR-HFn revealed that the addition of the MMPs recognition sequence did not affect the self-assembly of HFn but presented a surface-exposed loop susceptible to MMPs cleavage. Biochemical analysis indicated that this engineered protein cage is responsive to MMPs, enabling the targeted release of encapsulated drugs. To evaluate the therapeutic potential of this engineered protein cage, monosubstituted ß-carboxy phthalocyanine zinc (CPZ), a type of photosensitizer, was loaded inside this protein cage. The prepared CPZ@MR-HFn showed higher uptake and stronger phototoxicity in MMPs overexpressed tumor cells, as well as enhanced penetration into multicellular tumor spheroids compared with its counterpart CPZ@HFn in vitro. In vivo, CPZ@MR-HFn displayed a higher tumor inhibitory rate than CPZ@HFn under illumination. These results indicated that MR-HFn is a promising nanocarrier for anticancer drug delivery and the MMP-responsive strategy here can also be adapted for other stimuli.


Asunto(s)
Antineoplásicos , Liberación de Fármacos , Metaloproteinasas de la Matriz , Ingeniería de Proteínas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Metaloproteinasas de la Matriz/metabolismo , Animales , Línea Celular Tumoral , Ratones , Ferritinas/química , Ferritinas/metabolismo , Indoles/química , Indoles/farmacología , Portadores de Fármacos/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química
4.
Bioconjug Chem ; 35(5): 593-603, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38592684

RESUMEN

Ferritin is a multivalent, self-assembling protein scaffold found in most human cell types, in addition to being present in invertebrates, higher plants, fungi, and bacteria, that offers an attractive alternative to polymer-based drug delivery systems (DDS). In this study, the utility of the ferritin cage as a DDS was demonstrated within the context of T cell agonism for tumor killing. Members of the tumor necrosis factor receptor superfamily (TNFRSF) are attractive targets for the development of anticancer therapeutics. These receptors are endogenously activated by trimeric ligands that occur in transmembrane or soluble forms, and oligomerization and cell-surface anchoring have been shown to be essential aspects of the targeted agonism of this receptor class. Here, we demonstrated that the ferritin cage could be easily tailored for multivalent display of anti-OX40 antibody fragments on its surface and determined that these arrays are capable of pathway activation through cell-surface clustering. Together, these results confirm the utility, versatility, and developability of ferritin as a DDS.


Asunto(s)
Ferritinas , Humanos , Ferritinas/química , Ferritinas/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Sistemas de Liberación de Medicamentos
5.
ACS Nano ; 18(17): 11217-11233, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38627234

RESUMEN

Due to its intrinsic tumor-targeting attribute, limited immunogenicity, and cage architecture, ferritin emerges as a highly promising nanocarrier for targeted drug delivery. In the effort to develop ferritin cage-encapsulated cisplatin (CDDP) as a therapeutic agent, we found unexpectedly that the encapsulation led to inactivation of the drug. Guided by the structural information, we deciphered the interactions between ferritin cages and CDDP, and we proposed a potential mechanism responsible for attenuating the antitumor efficacy of CDDP encapsulated within the cage. Six platinum prodrugs were then designed to avoid the inactivation. The antitumor activities of these ferritin-platinum prodrug complexes were then evaluated in cells of esophageal squamous cell carcinoma (ESCC). Compared with free CDDP, the complexes were more effective in delivering and retaining platinum in the cells, leading to increased DNA damage and enhanced cytotoxic action. They also exhibited improved pharmacokinetics and stronger antitumor activities in mice bearing ESCC cell-derived xenografts as well as patient-derived xenografts. The successful encapsulation also illustrates the critical significance of comprehending the interactions between small molecular drugs and ferritin cages for the development of precision-engineered nanocarriers.


Asunto(s)
Antineoplásicos , Cisplatino , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Ferritinas , Profármacos , Profármacos/química , Profármacos/farmacología , Humanos , Ferritinas/química , Ferritinas/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/química , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Ratones , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Cisplatino/farmacología , Cisplatino/química , Diseño de Fármacos , Platino (Metal)/química , Platino (Metal)/farmacología , Ratones Desnudos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sistemas de Liberación de Medicamentos
6.
J Am Chem Soc ; 146(17): 11657-11668, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38641862

RESUMEN

All protein-directed syntheses of metal nanoclusters (NCs) and nanoparticles (NPs) have attracted considerable attention because protein scaffolds provide a unique metal coordination environment and can adjust the shape and morphology of NCs and NPs. However, the detailed formation mechanisms of NCs or NPs directed by protein templates remain unclear. In this study, by taking advantage of the ferritin nanocage as a biotemplate to monitor the growth of Fe-O NCs as a function of time, we synthesized a series of iron NCs with different sizes and shapes and subsequently solved their corresponding three-dimensional atomic-scale structures by X-ray protein crystallography and cryo-electron microscopy. The time-dependent structure analyses revealed the growth process of these Fe-O NCs with the 4-fold channel of ferritin as nucleation sites. To our knowledge, the newly biosynthesized Fe35O23Glu12 represents the largest Fe-O NCs with a definite atomic structure. This study contributes to our understanding of the formation mechanism of iron NCs and provides an effective method for metal NC synthesis.


Asunto(s)
Ferritinas , Tamaño de la Partícula , Ferritinas/química , Nanopartículas del Metal/química , Hierro/química , Modelos Moleculares , Cristalografía por Rayos X , Compuestos Férricos/química
7.
Biochem Biophys Res Commun ; 712-713: 149939, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38640729

RESUMEN

Human heavy-chain ferritin is a naturally occurring protein with high stability and multifunctionality in biological systems. This study aims to utilize a prokaryotic expression system to produce recombinant human heavy-chain ferritin nanoparticles and investigate their targeting ability in brain tissue. The human heavy-chain ferritin gene was cloned into the prokaryotic expression vector pET28a and transformed into Escherichia coli BL21 (DE3) competent cells to explore optimal expression conditions. The recombinant protein was then purified to evaluate its immunoreactivity and characteristics. Additionally, the distribution of the administered protein in normal mice and its permeability in an in vitro blood-brain barrier (BBB) model were measured. The results demonstrate that the purified protein can self-assemble extracellularly into nano-cage structures of approximately 10 nm and is recognized by corresponding antibodies. The protein effectively penetrates the blood-brain barrier and exhibits slow clearance in mouse brain tissue, showing excellent permeability in the in vitro BBB model. This study highlights the stable expression of recombinant human heavy-chain ferritin using the Escherichia coli prokaryotic expression system, characterized by favorable nano-cage structures and biological activity. Its exceptional brain tissue targeting and slow metabolism lay an experimental foundation for its application in neuropharmaceutical delivery and vaccine development fields.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Escherichia coli , Ferritinas , Nanopartículas , Proteínas Recombinantes , Animales , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ratones , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Nanopartículas/química , Ferritinas/metabolismo , Ferritinas/genética , Ferritinas/química , Apoferritinas/metabolismo , Apoferritinas/genética , Apoferritinas/química , Distribución Tisular
8.
J Agric Food Chem ; 72(13): 7464-7475, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38527235

RESUMEN

Ferritin is a cage-like protein with modifiable outer and inner surfaces. To functionalize ferritin with preferable carrier applications, caffeic acid was first covalently bound to the soybean ferritin outer surface to fabricate a caffeic acid-ferritin complex (CFRT) by alkali treatment (pH 9.0). A decreased content of free amino acid (0.34 µmol/mg) and increased polyphenol binding equivalent (63.76 nmol/mg) indicated the formation of CFRT (ferritin/caffeic acid, 1:80). Fluorescence and infrared spectra verified the binding of caffeic acids to the ferritin structure. DSC indicated that the covalent modification enhanced the thermal stability of CFRT. Besides, CFRT maintained the typically spherical shape of ferritin (12 nm) and a hydration radius of 7.58 nm. Moreover, the bioactive colorant betanin was encapsulated in CFRT to form betanin-loaded CFRT (CFRTB), with an encapsulation rate of 15.5% (w/w). The betanin stabilities in CFRTB were significantly improved after heat, light, and Fe3+ treatments, and its red color retention was enhanced relative to the free betanin. This study delves into the modifiable ferritin application as nanocarriers of dual molecules and gives guidelines for betanin as a food colorant.


Asunto(s)
Betacianinas , Ferritinas , Betacianinas/química , Ferritinas/química , Ácidos Cafeicos
9.
J Agric Food Chem ; 72(14): 7629-7654, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38518374

RESUMEN

Ferritin nanocages possess remarkable structural properties and biological functions, making them highly attractive for applications in functional materials and biomedicine. This comprehensive review presents an overview of the molecular characteristics, extraction and identification of ferritin, ferritin receptors, as well as the advancements in the directional design of high-order assemblies of ferritin and the applications based on its unique structural properties. Specifically, this Review focuses on the regulation of ferritin assembly from one to three dimensions, leveraging the symmetry of ferritin and modifications on key interfaces. Furthermore, it discusses targeted delivery of nutrition and drugs through facile loading and functional modification of ferritin. The aim of this Review is to inspire the design of micro/nano functional materials using ferritin and the development of nanodelivery vehicles for nutritional fortification and disease treatment.


Asunto(s)
Ferritinas , Ferritinas/química , Relación Estructura-Actividad
10.
Biomater Sci ; 12(9): 2408-2417, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38511491

RESUMEN

Alzheimer's disease is a severe brain condition caused by the formation of amyloid plaques composed of amyloid beta (Aß) peptides. These peptides form oligomers, protofibrils, and fibrils before deposition into amyloid plaques. Among these intermediates, Aß oligomers (AßOs) were found to be the most toxic and therefore an appealing target for drug development and understanding their role in the disease. However, precise isolation and characterization of AßOs have proven challenging because AßOs tend to aggregate and form heterogeneous mixtures in solution. As a solution, we genetically fused the Aß peptide with a ferritin monomer. Such fusion allowed the encapsulation of precisely 24 Aß peptides inside the 24-mer ferritin cage. Using high-speed atomic force microscopy (HS-AFM), we disassembled ferritin and directly visualized the Aß core enclosed within the cage. The thioflavin-T assay (ThT) and attenuated total reflection infrared spectroscopy (ATR-IR) revealed the presence of a ß-sheet structure in the encapsulated oligomeric aggregate. Gallic acid, an amyloid inhibitor, can inhibit the fluorescence of ThT bound AßOs. Our approach represents a significant advancement in the isolation and characterization of ß-sheet rich AßOs and is expected to be useful for future studies of other disordered peptides such as α-synuclein and tau.


Asunto(s)
Péptidos beta-Amiloides , Ferritinas , Conformación Proteica en Lámina beta , Péptidos beta-Amiloides/química , Ferritinas/química , Microscopía de Fuerza Atómica , Agregado de Proteínas/efectos de los fármacos , Humanos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación
11.
Angew Chem Int Ed Engl ; 63(16): e202401379, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38407997

RESUMEN

Ferritins are multimeric cage-forming proteins that play a crucial role in cellular iron homeostasis. All H-chain-type ferritins harbour a diiron site, the ferroxidase centre, at the centre of a 4 α-helical bundle, but bacterioferritins are unique in also binding 12 hemes per 24 meric assembly. The ferroxidase centre is known to be required for the rapid oxidation of Fe2+ during deposition of an immobilised ferric mineral core within the protein's hollow interior. In contrast, the heme of bacterioferritin is required for the efficient reduction of the mineral core during iron release, but has little effect on the rate of either oxidation or mineralisation of iron. Thus, the current view is that these two cofactors function in iron uptake and release, respectively, with no functional overlap. However, rapid electron transfer between the heme and ferroxidase centre of bacterioferritin from Escherichia coli was recently demonstrated, suggesting that the two cofactors may be functionally connected. Here we report absorbance and (magnetic) circular dichroism spectroscopies, together with in vitro assays of iron-release kinetics, which demonstrate that the ferroxidase centre plays an important role in the reductive mobilisation of the bacterioferritin mineral core, which is dependent on the heme-ferroxidase centre electron transfer pathway.


Asunto(s)
Ceruloplasmina , Hierro , Hierro/química , Ceruloplasmina/química , Escherichia coli/metabolismo , Ferritinas/química , Proteínas Bacterianas/metabolismo , Grupo Citocromo b/química , Minerales , Oxidación-Reducción , Hemo/metabolismo
12.
Adv Sci (Weinh) ; 11(17): e2309271, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38368258

RESUMEN

Well-defined nanostructures are crucial for precisely understanding nano-bio interactions. However, nanoparticles (NPs) fabricated through conventional synthesis approaches often lack poor controllability and reproducibility. Herein, a synthetic biology-based strategy is introduced to fabricate uniformly reproducible protein-based NPs, achieving precise control over heterogeneous components of the NPs. Specifically, a ferritin assembly toolbox system is developed that enables intracellular assembly of ferritin subunits/variants in Escherichia coli. Using this strategy, a proof-of-concept study is provided to explore the interplay between ligand density of NPs and their tumor targets/penetration. Various ferritin hybrid nanocages (FHn) containing human ferritin heavy chains (FH) and light chains are accurately assembled, leveraging their intrinsic binding with tumor cells and prolonged circulation time in blood, respectively. Further studies reveal that tumor cell uptake is FH density-dependent through active binding with transferrin receptor 1, whereas in vivo tumor accumulation and tissue penetration are found to be correlated to heterogeneous assembly of FHn and vascular permeability of tumors. Densities of 3.7 FH/100 nm2 on the nanoparticle surface exhibit the highest degree of tumor accumulation and penetration, particularly in tumors with high permeability compared to those with low permeability. This study underscores the significance of nanoparticle heterogeneity in determining particle fate in biological systems.


Asunto(s)
Ferritinas , Nanopartículas , Animales , Humanos , Ratones , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ferritinas/metabolismo , Ferritinas/química , Nanopartículas/química , Nanopartículas/metabolismo , Nanoestructuras/química , Neoplasias/metabolismo , Femenino , Ratones Endogámicos BALB C
13.
Inorg Chem ; 63(7): 3359-3365, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38315811

RESUMEN

The Fe(II) oxidation mechanism in the ferroxidase center of heavy chain ferritin has been studied extensively. However, the actual production of H2O2 was found to be substantially lower than expected at low flux of Fe(II) to ferritin subunits. Here, we demonstrated that H2O2 could interact with the di-iron nuclear center, leading to the production of hydroxyl radicals and oxygen. Two reaction intermediates were captured in the ferroxidase center by using the time-lapse crystallographic techniques in a shellfish ferritin. The crystal structures revealed the binding of H2O2 as a µ -1,2-peroxo-diferric species and the binding of O2 to the diferric structure. This investigation sheds light on the reaction between the di-iron nuclear center and H2O2 and provides insights for the exploitation of metalloenzymes.


Asunto(s)
Ferritinas , Hierro , Hierro/química , Ferritinas/química , Peróxido de Hidrógeno/química , Ceruloplasmina/química , Oxidación-Reducción , Compuestos Ferrosos/química
14.
Biomater Sci ; 12(5): 1249-1262, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38247338

RESUMEN

Ferritins are globular proteins with an internal cavity that enables the encapsulation of a plethora of low-mass compounds. Unfortunately, the overall negative surface charge of ferritin's internal cavity hampers efficient loading of negatively charged molecules. Therefore, we produced a genetically engineered human H-chain ferritin containing a cationic RKRK domain, reversing the natural net charge of the cavity to positive, thus allowing for efficient encapsulation of negatively charged siRNA. Due to the reversed, positive charge mediated by RKRK domains, the recombinant ferritin produced in E. coli inherently carries a load of bacterial RNA inside its cavity, turning the protein into an effective sponge possessing high affinity for DNA/RNA-binding substances that can be loaded with markedly higher efficiency compared to the wildtype protein. Using doxorubicin as payload, we show that due to its loading through the RNA sponge, doxorubicin is released in a sustained manner, with a cytotoxicity profile similar to the free drug. In summary, this is the first report demonstrating a ferritin/nucleic acid hybrid delivery vehicle with a broad spectrum of properties exploitable in various fields of biomedical applications.


Asunto(s)
Apoferritinas , ARN , Humanos , Apoferritinas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Ferritinas/genética , Ferritinas/química , Doxorrubicina/farmacología , Doxorrubicina/química
15.
Nat Commun ; 15(1): 233, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172125

RESUMEN

Biominerals, the inorganic minerals of organisms, are known mainly for their physical property-related functions in modern living organisms. Our recent discovery of the enzyme-like activities of nanomaterials, coined as nanozyme, inspires the hypothesis that nano-biominerals might function as enzyme-like catalyzers in cells. Here we report that the iron cores of biogenic ferritins act as natural nanozymes to scavenge superoxide radicals. Through analyzing eighteen representative ferritins from three living kingdoms, we find that the iron core of prokaryote ferritin possesses higher superoxide-diminishing activity than that of eukaryotes. Further investigation reveals that the differences in catalytic capability result from the iron/phosphate ratio changes in the iron core, which is mainly determined by the structures of ferritins. The phosphate in the iron core switches the iron core from single crystalline to amorphous iron phosphate-like structure, resulting in decreased affinity to the hydrogen proton of the ferrihydrite-like core that facilitates its reaction with superoxide in a manner different from that of ferric ions. Furthermore, overexpression of ferritins with high superoxide-diminishing activities in E. coli increases the resistance to superoxide, whereas bacterioferritin knockout or human ferritin knock-in diminishes free radical tolerance, highlighting the physiological antioxidant role of this type of nanozymes.


Asunto(s)
Escherichia coli , Superóxidos , Humanos , Escherichia coli/genética , Escherichia coli/metabolismo , Ferritinas/química , Hierro/metabolismo , Fosfatos
16.
Biomacromolecules ; 25(1): 177-187, 2024 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-38059469

RESUMEN

The precise assembly of multiple biomacromolecules into well-defined structures and materials is of great importance for various biomedical and nanobiotechnological applications. In this study, we investigate the assembly requirements for two-component materials using charged protein nanocages as building blocks. To achieve this, we designed several variants of ferritin nanocages to determine the surface characteristics necessary for the formation of large-scale binary three-dimensional (3D) assemblies. These nanocage variants were employed in protein crystallization experiments and macromolecular crystallography analyses, complemented by computational methods. Through the screening of nanocage variant combinations at various ionic strengths, we identified three essential features for successful assembly: (1) the presence of a favored crystal contact region, (2) the presence of a charged patch not involved in crystal contacts, and (3) sufficient distinctiveness between the nanocages. Surprisingly, the absence of noncrystal contact mediating patches had a detrimental effect on the assemblies, highlighting their unexpected importance. Intriguingly, we observed the formation of not only binary structures but also both negatively and positively charged unitary structures under previously exclusively binary conditions. Overall, our findings will inform future design strategies by providing some design rules, showcasing the utility of supercharging symmetric building blocks in facilitating the assembly of biomacromolecules into large-scale binary 3D assemblies.


Asunto(s)
Ferritinas , Sustancias Macromoleculares/química , Ferritinas/química , Cristalización
17.
Food Chem ; 439: 138132, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38081094

RESUMEN

The ferritin cage can not only load iron ions in its inner cavity, but also has the capacity to carry other metal ions, thus constructing a new biological nano-transport system. The nanoparticles formed by ferritin and minerals can be used as ingredients of mineral supplements, which overcome the shortcomings of traditional mineral ingredients such as low bioavailability. Moreover, ferritin can be used to remove heavy metal ions from contaminated food. Silver and palladium nanoparticles formed by ferritin are also applied as anticancer agents. Ferritin combined with metal ions can be also used to detect harmful substances. This review aims to provide a comprehensive overview of ferritin's function in transporting and binding metal ions, and discusses the limitations and future prospects, which offers valuable insights for the application of ferritin in mineral supplements, food detoxifiers, anticancer agents, and food detections.


Asunto(s)
Antineoplásicos , Nanopartículas del Metal , Ferritinas/química , Paladio , Minerales/metabolismo , Iones
18.
Biochim Biophys Acta Gen Subj ; 1868(2): 130525, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38043914

RESUMEN

The development of new drug delivery systems for targeted chemotherapy release in cancer cells represents a very promising tool. In this contest, protein-based nanocages have considerable potential as drug delivery devices. Notably, ferritin has emerged as an excellent candidate due to its unique architecture, surface properties and high biocompatibility. A promising strategy might then involve ferritin cargos for specifical release of AntiMicrobial Peptides endowed with anticancer activity to cancer cells. In this paper, we encapsulated the TRIL analogue of Temporin-L peptide within a ferritin nanocage and evaluated the cargo biological properties. The results demonstrated a reduced haemolytic activity of the peptide and a selective cytotoxicity activity on cancer cells likely mediated by oxidative stress while having no effects on non-tumoral cells. The combination of the properties of ferritin with TRIL, might open up the way to the development of novel peptide delivery systems for future pharmaceutical applications.


Asunto(s)
Ferritinas , Péptidos , Ferritinas/química , Péptidos/farmacología , Péptidos/química , Sistemas de Liberación de Medicamentos/métodos
19.
J Sci Food Agric ; 104(5): 2783-2791, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38009805

RESUMEN

BACKGROUND: Lycopene (LYC), a carotenoid found in abundance in ripe red fruits, exhibits higher singlet oxygen quenching activity than other carotenoids. However, the stability of LYC is extremely poor due to its high double-bond content. In this paper, a nano-encapsulation strategy based on highly stable marine-derived ferritin GF1 nanocages was used to improve the thermal stability and oxidation resistance of LYC, thereby boosting its functional effectiveness and industrial applicability. RESULTS: The preparation of GF1-LYC nanoparticles benefited from the pH-responsive reversible self-assembly of GF1 to capture LYC molecules into GF1 cavities with a LYC-to-protein ratio of 51 to 1. After the encapsulation of the LYC, the reassembled GF1 nanocages maintained intact morphology and good monodispersity. The GF1-LYC nanoparticles incorporated the characteristic LYC peaks in spectrograms, and their powder form contained the crystalline form of LYC. Molecular docking revealed that LYC bound with the inner triple-axis channel areas of GF1, interacting with VAL139, LYS72, LYS65, TYR69, PHE129, HIS133, HIS62, and TYR134 amino acids through hydrophobic bonds. Fourier transform infrared spectroscopy also demonstrated the bonding of GF1 and LYC. In comparison with free LYC, GF1 reduced the thermal degradation of encapsulated LYC at 37 °C significantly and maintained the 2,2-Diphenyl-1-picrylhydrazyl (DPPH)-scavenging ability of LYC. CONCLUSION: As expected, the water solubility, thermal stability, and antioxidant capacity of encapsulated LYC from GF1-LYC nanoparticles was notably improved in comparison with free LYC, indicating that the shell-like marine ferritin nanoplatform might enhance the stable delivery of LYC and promote its utilization in the field of food nutrition and in other industries. © 2023 Society of Chemical Industry.


Asunto(s)
Crassostrea , Ferritinas , Animales , Licopeno/metabolismo , Ferritinas/química , Simulación del Acoplamiento Molecular , Carotenoides/metabolismo
20.
Adv Healthc Mater ; 13(11): e2303623, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38142309

RESUMEN

PD-1/PD-L1 blockade immunotherapy has gained approval for the treatment of a diverse range of tumors; however, its efficacy is constrained by the insufficient infiltration of T lymphocytes into the tumor microenvironment, resulting in suboptimal patient responses. Here, a pioneering immunotherapy ferritin nanodrug delivery system denoted as ITFn-Pt(IV) is introduced. This system orchestrates a synergistic fusion of PD-L1 blockade, chemotherapy, and T-cell activation, aiming to augment the efficacy of tumor immunotherapy. Leveraging genetic engineering approach and temperature-regulated channel-based drug loading techniques, the architecture of this intelligent responsive system is refined. It is adept at facilitating the precise release of T-cell activating peptide Tα1 in the tumor milieu, leading to an elevation in T-cell proliferation and activation. The integration of PD-L1 nanobody KN035 ensures targeted engagement with tumor cells and mediates the intracellular delivery of the encapsulated Pt(IV) drugs, culminating in immunogenic cell death and the subsequent dendritic cell maturation. Employing esophageal squamous cell carcinoma (ESCC) as tumor model, the potent antitumor efficacy of ITFn-Pt(IV) is elucidated, underscored by augmented T-cell infiltration devoid of systemic adverse effects. These findings accentuate the potential of ITFn-Pt(IV) for ESCC treatment and its applicability to other malignancies resistant to established PD-1/PD-L1 blockade therapies.


Asunto(s)
Antígeno B7-H1 , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Linfocitos T , Animales , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/inmunología , Humanos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Ratones , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/inmunología , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/terapia , Línea Celular Tumoral , Activación de Linfocitos/efectos de los fármacos , Ferritinas/química , Microambiente Tumoral/efectos de los fármacos , Inmunoterapia/métodos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA