RESUMEN
It is known that the saturation ratio of transferrin (Tf) with iron in human blood is an important clinical parameter. Specific antibodies can be used to analyze subtle changes in the relative abundance of different forms of transferrin potentially associated with a pathological process. Recently, the authors of this study were able to obtain and characterize highly specific single-domain antibodies (nanobodies) that predominantly recognize the iron-saturated (holo-Tf) or iron-unsaturated (apo-Tf) form of transferrin. In this work, under conditions closer to physiological than in the previous experiments, we further demonstrated that these unique nanobodies have extremely high differential binding specificity for different forms of Tf in different human biological fluids. Using these nanobodies, we were able to analyze for the first time relative abundance of the transferrin forms in urine samples from the patients with bladder cancer (BC). We have shown that increase in the concentration of total Tf in the urine samples normalized for creatinine is associated with the degree of progress and growth of malignancy of BC. In the samples of healthy donors and in the early stages of BC (G1), Tf is detected in much smaller amounts (compared to the later stages) and only with additional concentration of the studied samples. For most of the studied urine samples from the BC patients, it is expected (as previously shown in the case of Tf in the blood of terminal ovarian cancer patients) that the concentration of apo-Tf is clearly higher than holo-Tf, especially in the case of the most advanced muscle-invasive BC. It was a surprise for us that approximately equal amounts of apo-Tf and holo-Tf were found in the urine samples of some patients with BC. We hypothesized that the holo-Tf fraction in this case could be largely represented by the "secondary complexes" formed by apo-Tf in combination with ions other than Fe3+, which accumulate in the urine of some cancer patients and are able to bind to apo-Tf, changing its conformation towards holo-Tf. By using inductively coupled plasma mass spectroscopy (ICP-MS), we obtained first results confirming our hypothesis. Preparation of the holo-Tf in these urine samples was found to be highly enriched in zinc and nickel. Also, relative enrichment in cadmium has been observed in this preparation, but at much lower concentrations. The obtained data indicate that the used nanobody, while recognizing predominantly the iron-saturated form of transferrin (holo-Tf), is also capable of binding transferrin in association with other metal ions that are different from iron. This ability could potentially open up new possibilities for investigation of relative abundance of various metal ions in association with transferrin in human biological fluids in normal and pathological conditions.
RESUMEN
Neutrophils apply several antimicrobial strategies including degranulation, phagocytosis, the generation of reactive oxygen species (ROS) and the release of neutrophil extracellular traps (NETs) to fight pathogens. Iron is considered to be an invaluable constituent of host immune defense and plays a dual role in immunity. It is a well-known component of antimicrobial proteins and is a necessary microelement for pathogen survival. The aim of this study was to broaden the knowledge regarding the impact of iron on the function of neutrophils. Neutrophils from healthy blood donors and patients with mild iron-deficiency anemia and HL-60 cells differentiated toward granulocyte-like cells were incubated with Fe2+ , Fe3+ or holo-transferrin (holo-Tf). Moreover, we isolated murine neutrophils of HFE gene knockout (KO) mice and mice fed iron-deficient, iron-equivalent and high-iron diets. We analyzed the release of NETs, phagocytosis, degranulation of azurophilic granules, ROS release, bactericidal activity of granulocytes against Escherichia coli and neutrophil elastase (NE) activity. We show that holo-Tf inhibits the release of NETs stimulated by phorbol 12-myristate 13-acetate by inhibiting NE activity. Studies performed in mice models reveal that iron overload inhibits the release of NETs and ROS production in neutrophils isolated from HFE KO mice and mice fed a high-iron diet. No impact of a low-iron diet on neutrophil phagocytosis, ROS production or release of NETs was observed. Our study underscores the physiological significance of iron in neutrophil function, specifically in the release of NETs.
Asunto(s)
Trampas Extracelulares , Sobrecarga de Hierro , Animales , Trampas Extracelulares/metabolismo , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Ratones , Neutrófilos , Especies Reactivas de Oxígeno/metabolismoRESUMEN
The blood-brain barrier (BBB) is the most important obstacle to improving the clinical outcomes of diagnosis and therapy of glioblastoma. Thus, the development of a novel nanoplatform that can efficiently traverse the BBB and achieve both precise diagnosis and therapy is of great importance. Herein, an intelligent nanoplatform based on holo-transferrin (holo-Tf) with in situ growth of MnO2 nanocrystals is constructed via a reformative mild biomineralization process. Furthermore, protoporphyrin (ppIX), acting as a sonosensitizer, is then conjugated into holo-Tf to obtain MnO2 @Tf-ppIX nanoparticles (TMP). Because of the functional inheritance of holo-Tf during fabrication, TMP can effectively traverse the BBB for highly specific magnetic resonance (MR) imaging of orthotopic glioblastoma. Clear suppression of tumor growth in a C6 tumor xenograft model is achieved via sonodynamic therapy. Importantly, the experiments also indicate that the TMP nanoplatform has satisfactory biocompatibility and biosafety, which favors potential clinical translation.
Asunto(s)
Barrera Hematoencefálica , Glioblastoma , Imagen por Resonancia Magnética , Nanocompuestos , Terapia por Ultrasonido , Animales , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioblastoma/diagnóstico por imagen , Glioblastoma/terapia , Imagen por Resonancia Magnética/métodos , Compuestos de Manganeso/química , Ratones , Ratones Desnudos , Óxidos/química , Terapia por Ultrasonido/métodosRESUMEN
Congenital toxoplasmosis is a serious health problem that can lead to miscarriage. HTR-8/SVneo is a first trimester extravillous trophoblast, while BeWo is a choriocarcinoma with properties of villous trophoblast cells. In the placenta, iron is taken up from Fe-transferrin through the transferrin receptor being the ion an important nutrient during pregnancy and also for Toxoplasma gondii proliferation. The aim of this study was to evaluate the role of iron in T. gondii proliferation in BeWo and HTR-8/SVneo cells and in human chorionic villous explants. The cells were infected with T. gondii, iron supplemented or deprived by holo-transferrin or deferoxamine, respectively, and parasite proliferation and genes related to iron balance were analyzed. It was verified that the addition of holo-transferrin increased, and DFO decreased the parasite multiplication in both trophoblastic cells, however, in a more expressive manner in HTR-8/SVneo, indicating that the parasite depends on iron storage in trophoblastic cells for its growth. Also, tachyzoites pretread with DFO proliferate normally in trophoblastic cells demonstrating that DFO itself does not interfere with parasite proliferation. Additionally, T. gondii infection induced enhancement in transferrin receptor mRNA expression levels in trophoblastic cells, and the expression was higher in HTR-8/SVneo compared with BeWo. Finally, DFO-treatment was able to reduce the parasite replication in villous explants. Thus, the iron supplementation can be a double-edged sword; in one hand, it could improve the supplement of an essential ion to embryo/fetus development, and on the other hand, could improve the parasite proliferation enhancing the risk of congenital infection.
Asunto(s)
Hierro/metabolismo , Complicaciones Infecciosas del Embarazo/parasitología , Toxoplasma/crecimiento & desarrollo , Toxoplasma/metabolismo , Toxoplasmosis/metabolismo , Trofoblastos/parasitología , Línea Celular Tumoral , Citoplasma/metabolismo , Femenino , Células HeLa , Humanos , Placenta/química , Placenta/parasitología , Embarazo , ARN Mensajero/biosíntesisRESUMEN
BACKGROUND: Transferrin and its receptors play an important role during the uptake and transcytosis of iron through blood-brain barrier (BBB) endothelial cells (ECs) to maintain iron homeostasis in BBB endothelium and brain. Any disruptions in the cell environment may change the distribution of transferrin receptors on the cell surface, which eventually alter the homeostasis and initiate neurodegenerative disorders. In this paper, we developed a comprehensive mathematical model that considers the necessary kinetics for holo-transferrin internalization and acidification, apo-transferrin recycling, and exocytosis of free iron and transferrin-bound iron through basolateral side of BBB ECs. METHODS: Ordinary differential equations are formulated based on the first order reaction kinetics to model the iron transport considering their interactions with transferrin and transferrin receptors. Unknown kinetics rate constants are determined from experimental data by applying a non-linear optimization technique. RESULTS: Using the estimated kinetic rate constants, the presented model can effectively reproduce the experimental data of iron transports through BBB ECs for many in-vitro studies. Model results also suggest that the BBB ECs can regulate the extent of the two possible iron transport pathways (free and transferrin-bound iron) by controlling the receptor expression, internalization of holo-transferrin-receptor complexes and acidification of holo-transferrin inside the cell endosomes. CONCLUSION: The comprehensive mathematical model described here can predict the iron transport through BBB ECs considering various possible routes from blood side to brain side. The model can also predict the transferrin and iron transport behavior in iron-enriched and iron-depleted cells, which has not been addressed in previous work.
Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Hierro/metabolismo , Modelos Cardiovasculares , Transporte Biológico Activo/fisiología , HumanosRESUMEN
Background and Aims: Liver iron overload can induce hepatic expression of bone morphogenic protein (BMP) 6 and activate the BMP/SMAD pathway. However, serum iron overload can also activate SMAD but does not induce BMP6 expression. Therefore, the mechanisms through which serum iron overload activates the BMP/SMAD pathway remain unclear. This study aimed to clarify the role of SMURF1 in serum iron overload and the BMP/SMAD pathway. Methods: A cell model of serum iron overload was established by treating hepatocytes with 2 mg/mL of holo-transferrin (Holo-Tf). A serum iron overload mouse model and a liver iron overload mouse model were established by intraperitoneally injecting 10 mg of Holo-Tf into C57BL/6 mice and administering a high-iron diet for 1 week followed by a low-iron diet for 2 days. Western blotting and real-time PCR were performed to evaluate the activation of the BMP/SMAD pathway and the expression of hepcidin. Results: Holo-Tf augmented the sensitivity and responsiveness of hepatocytes to BMP6. The E3 ubiquitin-protein ligase SMURF1 mediated Holo-Tf-induced SMAD1/5 activation and hepcidin expression; specifically, SMURF1 expression dramatically decreased when the serum iron concentration was increased. Additionally, the expression of SMURF1 substrates, which are important molecules involved in the transduction of BMP/SMAD signaling, was significantly upregulated. Furthermore, in vivo analyses confirmed that SMURF1 specifically regulated the BMP/SMAD pathway during serum iron overload. Conclusions: SMURF1 can specifically regulate the BMP/SMAD pathway by augmenting the responsiveness of hepatocytes to BMPs during serum iron overload.
RESUMEN
Recently, different alternative regulated cell death (RCD) pathways, viz., necroptosis, pyroptosis, ferroptosis, cuproptosis etc., have been explored as important targets for the development of cancer medications in recent years, as these can change the immunogenicity of the tumor microenvironment (TME) and will finally lead to the inhibition of cancer progression and metastasis. Here, we report the development of transferrin immobilized graphene oxide (Tfn@GOAPTES) nanocomposite as a therapeutic strategy toward cancer cell killing. The electrostatic immobilization of Tfn on the GOAPTES surface was confirmed by different spectroscopy and microscopy techniques. The Tfn immobilization was found to be â¼74 ± 4%, whereas the stability of the protein on the GO surface suggested a robust nature of the nanocomposite. The MTT assay suggested that Tfn@GOAPTES exhibited cytotoxicity toward HeLa cells via increased lipid peroxidation and DNA damage. Western blot studies resulted in decreased expression of acetylation on lysine 40 of α-tubulin and increased expression of LC3a/b for Tfn@GOAPTES treated HeLa cells, suggesting autophagy to be the main cause of the cell death mechanism. Overall, we predict that the present approach can be used as a therapeutic strategy for cancer cell killing via selective induction of a high concentration of intracellular iron.
Asunto(s)
Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Grafito , Nanocompuestos , Transferrina , Grafito/química , Grafito/farmacología , Humanos , Nanocompuestos/química , Transferrina/química , Transferrina/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Células HeLa , Tamaño de la Partícula , Ensayo de Materiales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Hierro/química , Hierro/metabolismo , Supervivencia Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacosRESUMEN
In this work, the interaction of human serum albumin (HSA) and human holo-transferrin (HTF) with the prepared Nano-Kaempferol (Nano-KMP) through oil-in-water procedure was investigated in the form of binary and ternary systems by the utilization of different spectroscopy techniques along with molecular simulation and cancer cell experiments. According to fluorescence spectroscopy outcomes, Nano-KMP is capable of quenching both proteins as binary systems by a static mechanism, while in the form of (HSA-HTF) Nano-KMP as the ternary system, an unlinear Stern-Volmer plot was elucidated with the occurrence of both dynamic and static fluorescence quenching mechanisms in the binding interaction. In addition, the two acquired Ksv values in the ternary system signified the existence of two sets of binding sites with two different interaction behaviors. The binding constant values of HSA-Nano KMP, HTF-Nano-KMP, and (HSA-HTF) Nano-KMP complexes formation were (2.54 ± 0.03) × 104, (2.15 ± 0.02) × 104 and (1.43 ± 0.04) × 104M-1at the first set of binding sites and (4.68 ± 0.05) × 104 M-1 at the second set of binding sites, respectively. The data of thermodynamic parameters confirmed the major roles of hydrogen binding and van der Waals forces in the formation of HSA-Nano KMP and HTF-Nano KMP complexes. The thermodynamic parameter values of (HSA-HTF) Nano KMP revealed the dominance of hydrogen binding and van der Waals forces in the first set of binding sites and hydrophobic forces for the second set of binding sites. Resonance light scattering (RLS) analysis displayed the existence of a different interaction behavior for HSA-HTF complex in the presence of Nano-KMP as the ternary system. Moreover, circular dichroism (CD) technique affirmed the conformational changes of the secondary structure of proteins as binary and ternary systems. Molecular docking and molecular dynamics simulations (for 100 ns) were performed to investigate the mechanism of KMP binding to HSA, HTF, and HSA-HTF. Next to observing a concentration and time-dependent cytotoxicity, the down regulation of PI3K/AkT/mTOR pathway resulted in cell cycle arrest in SW480 cells.
Asunto(s)
Fosfatidilinositol 3-Quinasas , Albúmina Sérica Humana , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , Fosfatidilinositol 3-Quinasas/metabolismo , Espectrometría de Fluorescencia , Sitios de Unión , Dicroismo Circular , Albúmina Sérica Humana/química , Termodinámica , Transferrina/química , Hidrógeno , Agua/metabolismoRESUMEN
The intracellular redox-active labile iron pool (LIP) is weakly chelated and available for integration into the iron metalloproteins that are involved in diverse cellular processes, including cancer cell-specific metabolic oxidative stress. Abnormal iron metabolism and elevated LIP levels are linked to the poor survival of lung cancer patients, yet the underlying mechanisms remain unclear. Depletion of the LIP in non-small-cell lung cancer cell lines using the doxycycline-inducible overexpression of the ferritin heavy chain (Ft-H) (H1299 and H292), or treatment with deferoxamine (DFO) (H1299 and A549), inhibited cell growth and decreased clonogenic survival. The Ft-H overexpression-induced inhibition of H1299 and H292 cell growth was also accompanied by a significant delay in transit through the S-phase. In addition, both Ft-H overexpression and DFO in H1299 resulted in increased single- and double-strand DNA breaks, supporting the involvement of replication stress in the response to LIP depletion. The Ft-H and DFO treatment also sensitized H1299 to VE-821, an inhibitor of ataxia telangiectasis and Rad2-related (ATR) kinase, highlighting the potential of LIP depletion, combined with DNA damage response modifiers, to alter lung cancer cell responses. In contrast, only DFO treatment effectively reduced the LIP, clonogenic survival, cell growth, and sensitivity to VE-821 in A549 non-small-cell lung cancer cells. Importantly, the Ft-H and DFO sensitized both H1299 and A549 to chemoradiation in vitro, and Ft-H overexpression increased the efficacy of chemoradiation in vivo in H1299. These results support the hypothesis that the depletion of the LIP can induce genomic instability, cell death, and potentiate therapeutic responses to chemoradiation in NSCLC.
RESUMEN
In this work, we investigated the simultaneous binding of curcumin (CUR) to human serum albumin (HSA) and human-holo transferrin (HTF) in the roles of binary and ternary systems. The binding affinity and binding site of protein-protein interaction were studied by the methods of multiple spectroscopic and molecular dynamics (MD) simulation. According to the results, the measurements for binding constant of HSA-CUR, HTF-CUR and (HSA-HTF) CUR complexes were observed to be 1.51 × 105, 7.93 × 104 and 1.44 × 105 M-1 respectively. Thermodynamic parameters were considered to be set at three varying temperatures including 298, 303, and 308 K. In conformity to the negative values of ΔH0 and ΔS0 the significant roles of hydrogen binding and van der-Waals forces in the formation of complexes are quiet evident. The binding distance between Trp residues of HSA, HTF and HSA-HTF upon interaction with CUR, were acquired by applying the Förster's theory of non-radioactive energy transfer and reported to be 2.04 nm, 1.78 nm, and 1.86 nm, respectively. In accordance with the conductometry and Resonance light scattering (RLS) results, there were different interaction behaviors among the HSA-HTF complex and CUR in ternary system when being compared to the outcomes of binary system. The secondary structure of all three cases increased as the CUR concentration was intensified, which confirmed the inducement of proteins conformational changes through the application of circular dichroism (CD) technique. The experimental results that were acquired throughout the binding of HSA-CUR, HTF-CUR, and (HSA-HTF) CUR complexes were approved by molecular modeling.Communicated by Ramaswamy H. Sarma.
Asunto(s)
Curcumina , Albúmina Sérica Humana , Humanos , Albúmina Sérica Humana/química , Simulación de Dinámica Molecular , Transferrina/química , Unión Proteica , Dicroismo Circular , Sitios de Unión , Termodinámica , Espectrometría de Fluorescencia/métodos , Simulación del Acoplamiento MolecularRESUMEN
A highly efficient technology for generating new monoclonal single-domain recombinant antibodies (nanobodies) was used to obtain a panel of nanobodies recognizing human apo- and/or holo-transferrin. This article is devoted to the primary analysis of the properties of two different variants of the new nanobodies obtained by us, as well as to the demonstration of the unique potential of their application for diagnostic studies. The simultaneous use of immunosorbents based on these nanobodies apparently makes it possible to detect changes in the relative abundance of apo- and holo-transferrin in human biological fluids. Such changes could potentially be indicative of an increased risk or degree of development of pathological processes, such as malignant neoplasms in humans.
RESUMEN
(1) Background: There are no reliable and widely available markers of functional iron deficiency (FID) in cancer. The aim of the study was to evaluate the role of transferrin (Tf) as a marker of cancer of the ovary (CrO) and related FID. (2) Methods: The study groups consisted of 118 patients with CrO and 69 control females. Blood serum iron status was determined on a Beckman Coulter AU (USA) analyzer. Tf quantification was performed by immunoturbidimetry. The relative contents of apo- and holo-Tf (iron-free and iron-saturated Tf, respectively) were determined in eight patients and a control female by immunochromatographic analysis based on the use of monoclonal single-domain antibodies (nanobodies). (3) Results: Four groups of patients with different iron statuses were selected according to ferritin and transferrin saturation values: absolute iron deficiency (AID) (n = 42), FID (n = 70), iron overload (n = 4), normal iron status (n = 2). The groups differed significantly in Tf values (p < 0.0001). Lower values of Tf were associated with FID. Furthermore, FID is already found in the initial stages of CrO (26%). Immunosorbents based on nanobodies revealed the accumulation of apo-Tf and the decrease in holo-Tf in patients with CrO. (4) Conclusions: Tf may be a promising tool for diagnosing both CrO and associated FID.
RESUMEN
Extraintestinal pathogenic Escherichia coli (ExPEC) is a common anthropozoonotic pathogen that causes systemic infections. To establish infection, ExPEC must utilize essential nutrients including iron from the host. Transferrin is an important iron source for multiple bacteria. However, the mechanism by which ExPEC utilizes transferrin remains unclear. In this study, we found that iron-saturated holo-transferrin rather than iron-free apo-transferrin promoted the vitality of ExPEC in heat-inactivated human serum. The multifunctional protein Elongation factor Tu (EFTu) worked as a holo-transferrin binding protein. EFTu not only bound holo-transferrin rather than apo-transferrin but also released transferrin-related iron, with all domains of EFTu involved in holo-transferrin binding and iron release events. We also identified the surface location of EFTu on ExPEC. Overexpression of EFTu on the surface of nonpathogenic E. coli not only promoted the binding of bacteria to holo-transferrin but also facilitated the uptake of transferrin-related iron. More importantly, it significantly enhanced the survival of E. coli in heat-inactivated human serum, which was positively correlated with holo-transferrin but not apo-transferrin. Our research revealed a novel function of EFTu in binding holo-transferrin to promote iron uptake by bacteria, suggesting that EFTu was a potential virulence factor of ExPEC. In addition, our study provided research avenues into the iron acquisition and pathogenicity mechanisms of ExPEC.
Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Escherichia coli Patógena Extraintestinal , Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Hierro/metabolismo , Factor Tu de Elongación Peptídica/genética , Factor Tu de Elongación Peptídica/metabolismo , TransferrinaRESUMEN
The interactions of proteins with drugs are very important from a pharmacological point of view. Holo-transferrin is a blood-plasma glycoprotein whose main function is iron-binding and the transport of other ligands. Additionally, the protein is only transferrin-form recognized by TfR1 and TfR2 receptors at the surface of rapidly proliferating malignant cells. Imatinib mesylate is a tyrosine-kinase inhibitor mainly used in the treatment of blood cancers, frequently in multidrug therapy with cyclophosphamide. In this study the effect of cyclophosphamide on the interaction of imatinib mesylate with human holo-transferrin has been investigated. Using spectroscopic techniques such as fluorescence, circular dichroism, ultraviolet-visible and electrophoretic light scattering additive parameters, system stability and the effect of the ligands on the protein conformation at varying pH values have been defined. Calculated quenching constants are in the order of 2 × 104 M-1 and the type of interaction depends on the reaction medium. Under physiological conditions binding constant is 1.329 × 106 M-1 whereas in an environment similar to that of cancer cells the constant is significantly lower, Ka = 6.060 × 104 M-1. N values are approximate to 1 in all cases. Moreover, some changes are observed in the α-helical structure of the protein after interaction with the drugs and the presence of cyclophosphamide slightly stabilizes the protein secondary structure. All collected data proves the effect of cyclophosphamide on the interaction between imatinib mesylate and human holo-transferrin. It is of great clinical interest due to anticancer, multidrug therapies including both imatinib mesylate and cyclophosphamide.
Asunto(s)
Leprostáticos , Transferrina , Ciclofosfamida , Quimioterapia Combinada , Humanos , Mesilato de Imatinib , Unión Proteica , Transferrina/metabolismoRESUMEN
In this study, we have investigated the effects of Nano-curcumin (Nano-CUR) binding on HSA-HTF interactions as binary and ternary systems, which had been done through multiple spectroscopic and MD simulation. It has been indicated by fluorescence spectroscopy that Nano-CUR is capable of quenching both proteins with a static mechanism. Thermodynamic parameters have been calculated by considering the fluorescence data at different temperatures. The binding constants of HSA-Nano-CUR, HTF-Nano-CUR and (HSA-HTF) Nano-CUR complexes formation were (2.03 ± 0.32)×107 M-1, (2.46 ± 0.32)×106 and (4.54 ± 0.32)×106 M-1 respectively. According to the negative values of ΔH0 and ΔS0, the roles of van-der-Waals forces and hydrogen bond are quite essential throughout this particular binding. Besides, the negative ΔH0 and ΔS0 values of HTF (Nano-CUR) have been larger than those of the HSA (Nano-CUR) and HSA-HTF (Nano-CUR), which demonstrates the higher significance of interaction bonding. As it had been detected through the synchronized fluorescence spectroscopy at Δλ = 60 nm, the position of Nano-CUR with mixed protein in ternary system has been closer to Tyr residues. Relatively, the binding distances between Trp residues of HSA and HTF in HSA (Nano-CUR), HTF (Nano-CUR), and (HSA-HTF Nano-CUR) complexes, which had been procured in accordance with the fluorescence resonance energy transfer (FRET), have been found to be 1.82 nm, 1.87 nm, and 1.92 nm, respectively. We have evaluated the induced conformational changes of two proteins throughout the binding of Nano-CUR to HSA and HTF as binary and ternary systems by employing the CD technique, while the formation of self-assemblies has been studied through MD simulation.
Asunto(s)
Curcumina , Sitios de Unión , Proteínas Sanguíneas , Dicroismo Circular , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , Espectrometría de Fluorescencia , Termodinámica , TransferrinaRESUMEN
Berberine is widely used in traditional Iranian medicine to treat diabetes and inflammatory conditions. This study was aimed at developing a method for the preparation of Berberine nanoparticles (Nano-Ber) in order to improve its aqueous-phase solubility and its complex formation with human serum albumin (HSA) and holo-transferrin (HTF) from the viewpoint of interaction behavior. Nano-Ber was prepared with olive oil as the oil phase, Tween 80 as the surfactant and Span 60 as the co-surfactant. Nano-Ber was obtained with a spherical shape and a mean particle size of 43.7 ± 3.6 nm, with an optimal oil:surfactant:co-surfactant ratio of 1:2:2, w/w/w. The antioxidant activity of Nano-Ber in comparison with Berberine was tested using DPPH and it was found that Nano-Ber had a large antioxidant activity. The cytotoxicity effects of Nano-Ber and Berberine on HepG2 were compared by MTT assay and detected in the treated HepG2 cells at concentrations up to 0.1 mM. The binding constants of HSA-Nano-Ber and HTF-Nano-Ber complexes formation were (2.93 ± 0.02) × 104 and (9.62 ± 0.03) × 103 M -1, respectively. Hydrogen bonds and van der Waals interactions were the predominant forces in the HSA-Nano-Ber and HTF-Nano-Ber complexes, and the process of Nano-Ber binding HSA and HTF was driven by ΔH 0 = -122.76 kJ mol-1, ΔS 0 = -325.49 J mol-1K-1 for HSA and ΔH 0 = -125.09 kJ mol-1, ΔS 0 = -43.37 J mol-1K-1 for HTF. The results of the simulation demonstrated that the Nano-Ber molecules were stabilized on the surface of final aggregates through both hydrophilic and hydrophobic interactions. Communicated by Ramaswamy Sarma.
Asunto(s)
Berberina , Albúmina Sérica Humana , Sitios de Unión , Humanos , Irán , Aceite de Oliva , Unión Proteica , Espectrometría de Fluorescencia , Termodinámica , Transferrina , AguaRESUMEN
Receptor heterogeneity in cancer is a major limitation of molecular targeting for cancer therapeutics. Single-receptor-targeted treatment exerts selection pressures that result in treatment escape for low-receptor-expressing tumor subpopulations. To overcome this potential for heterogeneity-driven resistance to molecular targeted photodynamic therapy (PDT), we present for the first time a triple-receptor-targeted photoimmuno-nanoconjugate (TR-PIN) platform. TR-PIN functionalization with cetuximab, holo-transferrin, and trastuzumab conferred specificity for epidermal growth factor receptor (EGFR), transferrin receptor (TfR), and human epidermal growth factor receptor 2 (HER-2), respectively. The TR-PINs exhibited up to a 24-fold improvement in cancer cell binding compared with EGFR-specific cetuximab-targeted PINs (Cet-PINs) in low-EGFR-expressing cell lines. Photodestruction using TR-PINs was significantly higher than the monotargeted Cet-PINs in heterocellular 3D in vitro models of heterogeneous pancreatic ductal adenocarcinoma (PDAC; MIA PaCa-2 cells) and heterogeneous head and neck squamous cell carcinoma (HNSCC, SCC9 cells) containing low-EGFR-expressing T47D (high TfR) or SKOV-3 (high HER-2) cells. Through their capacity for multiple tumor target recognition, TR-PINs can serve as a unique and amenable platform for the effective photodynamic eradication of diverse tumor subpopulations in heterogeneous cancers to mitigate escape for more complete and durable treatment responses.
RESUMEN
The interactions between estradiol and two carrier proteins, i.e. human serum albumin (HSA) and holo-transferrin (HTF) in aqueous solution at pH = 7.4 were studied by three-dimensional fluorescence emission spectroscopy, isothermal titration calorimetry (ITC), zeta-potential, resonance light-scattering and molecular modeling. Extensive fluorescence quenching was observed throughout the interaction between the drug and both proteins. Moreover, conformational changes were determined by observing the rearrangement of Trp residues during binding of estradiol with HSA and HTF at different concentrations. ITC experiments revealed that, in the presence of estradiol, both van der Waals forces and hydrogen bonding became predominant. In addition, other binding parameters such as enthalpy and entropy changes were determined by the zeta potential method. Molecular modeling suggested that estradiol was situated within sub-domain IB sited in the hydrophobic cluster in Site I, whereas the drug was located in the N-terminal of HTF where it was hydrogen bonded with Ala 670.
Asunto(s)
Estradiol/química , Albúmina Sérica Humana/química , Transferrina/química , Sitios de Unión , Calorimetría/métodos , Dicroismo Circular/métodos , Entropía , Fluorescencia , Transferencia Resonante de Energía de Fluorescencia/métodos , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Unión Proteica , Dominios Proteicos , Espectrometría de Fluorescencia/métodos , TermodinámicaRESUMEN
Active-targeted cancer imaging and therapy of glioma has attracted much attention in theranostic nanomedicine. As a promising tumor-targeting ligand, holo-transferrin (holo-Tf) has been applied for enhancing delivery of nanotheranostics. However, holo-Tf-based nanoassemblies for active targeting mediated multimodal imaging and therapeutics have not been previously reported. Here, we develop a one-step method for the preparation of holo-Tf-indocyanine green (holo-Tf-ICG) nanoassemblies for fluorescence (FL) and photoacoustic (PA) dual-modal imaging and photothermal therapy (PTT) of glioma. The nanoassemblies are formed by hydrophobic interaction and hydrogen bonds between holo-Tf and ICG, which exhibit excellent active tumor-targeting and high biocompability. The brain tumor with highly expressed Tf receptor can be clearly observed with holo-Tf-ICG nanoassemblies base on FL and PA dual-modal imaging in subcutaneous and orthotopic glioma models. Under the near-infrared laser irradiation, the holo-Tf-ICG nanoassemblies accumulated in tumor regions can efficiently convert laser energy into hyperthermia for tumor ablation. The novel theranostic nanoplatform holds great promise for precision diagnosis and treatment of glioma.
Asunto(s)
Verde de Indocianina/química , Glioma , Humanos , Nanopartículas , Nanomedicina Teranóstica , TransferrinaRESUMEN
The purpose of this study was to determine how lomefloxacin (LMF) interacts with human holo-transferrin (HTF) in the presence of two kinds of essential and nonessential amino acids. The investigations were carried out by fluorescence spectroscopy, zeta potential and molecular modeling techniques under imitated physiological conditions. We were able to determine the number of binding sites, the drug binding affinity to HTF in the presence of essential and nonessential amino acids and the quenching source of HTF. The interaction between HTF with LMF suggested that the microenvironment of the Trp residues was altered causing a strong static fluorescence quenching in the binary and ternary systems. The results pointed at the formation of a complex in the binary and ternary systems which caused an enhancement of the RLS intensity that was analyzed using synchronous fluorescence spectroscopy. The density functional theory (DFT) was employed to determine the amino acid residues on HTF that interacted with LMF. Also, Steric and van der Waals forces as well as the contribution of small amounts of hydrogen bonds were stronger or Tyr 71 in chain (b) than for 128 Trp in chain (a) of HTF.