Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 296
1.
J Mol Biol ; 435(20): 168262, 2023 10 15.
Article En | MEDLINE | ID: mdl-37678707

Transferrin receptor 1 (TfR) delivers iron across cellular membranes by shuttling the ion carrier protein transferrin. This ability to deliver large protein ligands inside cells is taken advantage of by pathogens to infiltrate human cells. Notably, the receptor's outermost ectodomain, the apical domain, is used as a point of attachment for several viruses including hemorrhagic arenaviruses. To better understand interactions with the receptor it would be advantageous to probe sequence determinants in the apical domain with viral spike proteins. Here, we carried out affinity maturation of our computationally designed apical domain from human TfR to identify underlying driving forces that lead to better binding. The improved variants were confirmed by in vitro surface plasmon resonance measurements with dissociation constants obtained in the lower nanomolar range. It was found that the strong binding affinities for the optimized variants matched the strength of interactions with the native receptor. The structure of the best variant was determined experimentally indicating that the conformational change in the hairpin binding motif at the protein-protein interface plays a crucial role. The experimental methodology can be straightforwardly applied to other arenavirus or pathogens that use the apical domain. It can further be useful to probe host-virus compatibility or therapeutic strategies based on the transferrin receptor decoys.


Arenaviruses, New World , Host-Pathogen Interactions , Receptors, Transferrin , Humans , Arenaviruses, New World/metabolism , Glycoproteins/chemistry , Protein Binding , Receptors, Transferrin/chemistry , Transferrin/chemistry , Transferrin/metabolism , Viral Proteins/metabolism
2.
Chembiochem ; 24(10): e202200795, 2023 05 16.
Article En | MEDLINE | ID: mdl-37005222

The transferrin receptor (TfR) mediates transcytosis across the blood-brain barrier (BBB), which offers a promising approach for the non-invasive delivery of therapeutics into the brain parenchyma. Employing the recombinant homodimeric murine TfR ectodomain, prepared in a biochemically functional state, we have selected a cognate Anticalin via phage display and bacterial cell surface display from a random library based on the human lipocalin 2 (Lcn2). After affinity maturation, several engineered lipocalin variants were identified that bind murine TfR in a non-competitive manner with the natural ligand (transferrin ⋅ Fe3+ ), among those an Anticalin - dubbed FerryCalin - exhibiting a dissociation constant (KD ) of 3.8 nM. Epitope analysis using the SPOT technique revealed a sequential epitope in a surface region of TfR remote from the transferrin-binding site. Due to the fast kon rate and short complex half-life, as evidenced by real-time surface plasmon resonance (SPR) measurements, FerryCalin, or one of its related mutants, shows characteristics as a potential vehicle for the brain delivery of biopharmaceuticals.


Lipocalins , Receptors, Transferrin , Mice , Humans , Animals , Lipocalins/genetics , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Brain/metabolism , Transferrin/chemistry , Transferrin/metabolism , Epitopes
3.
Inorg Chem ; 62(2): 675-678, 2023 Jan 16.
Article En | MEDLINE | ID: mdl-36602395

The molecular mechanism of how human serum transferrin (hTF) recognizes cisplatin at the atomic level is still unclear. Here, we report the molecular structure of the adduct formed upon the reaction of hTF with cisplatin. Pt binds the side chain of Met256 (at the N-lobe), without altering the protein overall conformation.


Cisplatin , Transferrin , Humans , Cisplatin/metabolism , Transferrin/chemistry , Iron/chemistry , Protein Conformation , Protein Binding , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism
4.
Nano Lett ; 22(15): 6328-6333, 2022 08 10.
Article En | MEDLINE | ID: mdl-35900277

DNA-assembled multiaptamer systems have been demonstrated to significantly promote the aptamer capacity of binding cell-surface-expressed proteins. However, how to conveniently harness them for efficient transmembrane delivery of targets remains a challenge. Toward this goal, here we engineer a G-quadruplex-proximized aptamer (G4PA) system in which a DNA aptamer specific for transferrin receptor (TfR) is guided by a bimolecular G4 and assembles into a dimerized proximity form that well matches homodimeric TfR highly expressed on the cancer cell surface. This system displays a higher capacity for targeting cell-surface TfR than the monomeric aptamer and super transmembrane transportation of nucleic acid cargoes, which is comparable to that of conventional liposome transfection but overcomes the lack of targeting ability of the latter. The G4PA system is then applied to the targeted delivery of siRNA for PLK1 gene silencing in positive cells rather than negative controls, showing great promise for use in precise anticancer therapy.


Aptamers, Nucleotide , Transferrin , Aptamers, Nucleotide/genetics , Liposomes , RNA, Small Interfering/genetics , Receptors, Cell Surface/metabolism , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Transferrin/chemistry
5.
FEBS J ; 289(10): 2935-2947, 2022 05.
Article En | MEDLINE | ID: mdl-34862739

Human transferrin receptor 1 (TfR) is necessary for the delivery of the iron carrier protein transferrin into cells and can be utilized for targeted delivery across cellular membranes. Binding of transferrin to the receptor is regulated by hereditary hemochromatosis protein (HFE), an iron regulatory protein that partly shares a binding site with transferrin on TfR. Here, we derived essential binding interactions from HFE and computationally grafted these into a library of small protein scaffolds. One of the designed proteins, TB08, was further optimized computationally and experimentally to identify variants with improved binding to TfR. The optimized variant, TB08 S3.1, expressed well in the E. coli expression system and had an affinity to TfR in the low micromolar range, Kd ≈ 1 µm, as determined by surface plasmon resonance. A binding competition assay with transferrin further confirmed the interaction of the evolved variant to TfR at the shared binding surface. Additionally, the GFP-tagged evolved variant of TB08 demonstrated cellular internalization as determined by fluorescent and confocal microscopy in HeLa cells. The designed protein is small, allows for robust cargo tagging, and interacts specifically with TfR, thus making it a valuable tool for the characterization of TfR-mediated cellular transport mechanisms and for the assessment of engineering strategies for cargo delivery across cell membranes.


Receptors, Transferrin , HeLa Cells , Humans , Membrane Proteins/metabolism , Protein Domains , Protein Engineering , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics , Transferrin/chemistry
6.
J Virol ; 95(17): e0186820, 2021 08 10.
Article En | MEDLINE | ID: mdl-34132574

Pathogenic clade B New World mammarenaviruses (NWM) can cause Argentine, Venezuelan, Brazilian, and Bolivian hemorrhagic fevers. Sequence variability among NWM glycoproteins (GP) poses a challenge to the development of broadly neutralizing therapeutics against the entire clade of viruses. However, blockade of their shared binding site on the apical domain of human transferrin receptor 1 (hTfR1/CD71) presents an opportunity for the development of effective and broadly neutralizing therapeutics. Here, we demonstrate that the murine monoclonal antibody OKT9, which targets the apical domain of hTfR1, can sterically block cellular entry by viral particles presenting clade B NWM glycoproteins (GP1-GP2). OKT9 blockade is also effective against viral particles pseudotyped with glycoproteins of a recently identified pathogenic Sabia-like virus. With nanomolar affinity for hTfR1, the OKT9 antigen binding fragment (OKT9-Fab) sterically blocks clade B NWM-GP1s and reduces infectivity of an attenuated strain of Junin virus. Binding of OKT9 to the hTfR1 ectodomain in its soluble, dimeric state produces stable assemblies that are observable by negative-stain electron microscopy. A model of the OKT9-sTfR1 complex, informed by the known crystallographic structure of sTfR1 and a newly determined structure of the OKT9 antigen binding fragment (Fab), suggests that OKT9 and the Machupo virus GP1 share a binding site on the hTfR1 apical domain. The structural basis for this interaction presents a framework for the design and development of high-affinity, broadly acting agents targeting clade B NWMs. IMPORTANCE Pathogenic clade B NWMs cause grave infectious diseases, the South American hemorrhagic fevers. Their etiological agents are Junin (JUNV), Guanarito (GTOV), Sabiá (SABV), Machupo (MACV), Chapare (CHAV), and a new Sabiá-like (SABV-L) virus recently identified in Brazil. These are priority A pathogens due to their high infectivity and mortality, their potential for person-to-person transmission, and the limited availability of effective therapeutics and vaccines to curb their effects. While low homology between surface glycoproteins of NWMs foils efforts to develop broadly neutralizing therapies targeting NWMs, this work provides structural evidence that OKT9, a monoclonal antibody targeting a single NWM glycoprotein binding site on hTfR1, can efficiently prevent their entry into cells.


Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Arenaviruses, New World/physiology , Glycoproteins/immunology , Hemorrhagic Fever, American/prevention & control , Receptors, Transferrin/immunology , A549 Cells , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , Humans , Protein Structure, Tertiary , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics
7.
Mol Biochem Parasitol ; 242: 111361, 2021 03.
Article En | MEDLINE | ID: mdl-33450336

The transferrin receptor of Trypanosoma brucei (TbTfR) is a heterodimer of a glycosylphosphatidylinositol (GPI)-anchored ESAG6 subunit and an ESAG7 subunit. To investigate whether the GPI-anchor is essential for the function of the TbTfR, an ESAG6 with a transmembrane domain instead of a GPI-anchor (ESAG6tmd) was inducibly expressed in bloodstream form trypanosomes. It is shown that the ESAG6tmd is able to dimerise with ESAG7 to form a TbTfR that can bind transferrin. Fractionation experiments clearly demonstrated that the transmembrane-anchored TbTfR is exclusively associated with the membrane fraction. No difference in the uptake of transferrin was observed between trypanosomes inducibly expressing a transmembrane-anchored TbTfR and trypanosomes inducibly expressing a GPI-anchored TbTfR. Differences in glycosylation pattern of ESAG6tmd and native ESAG6 may indicate different intracellular trafficking of transmembrane- and GPI-anchored TbTfRs. The findings suggest that the GPI-anchor is not essential for the function of the TbTfR in bloodstream forms of T. brucei.


Glycosylphosphatidylinositols/metabolism , Protein Processing, Post-Translational , Protein Subunits/metabolism , Protozoan Proteins/metabolism , Receptors, Transferrin/metabolism , Trypanosoma brucei brucei/metabolism , Base Sequence , Cell Membrane/chemistry , Cell Membrane/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Glycosylation , Glycosylphosphatidylinositols/chemistry , Plasmids/chemistry , Plasmids/metabolism , Protein Domains , Protein Engineering , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trypanosoma brucei brucei/genetics
8.
Proteins ; 89(3): 311-321, 2021 03.
Article En | MEDLINE | ID: mdl-33068039

Machupo virus, known to cause hemorrhagic fevers, enters human cells via binding with its envelope glycoprotein to transferrin receptor 1 (TfR). Similarly, the receptor interactions have been explored in biotechnological applications as a molecular system to ferry therapeutics across the cellular membranes and through the impenetrable blood-brain barrier that effectively blocks any such delivery into the brain. Study of the experimental structure of Machupo virus glycoprotein 1 (MGP1) in complex with TfR and glycoprotein sequence homology has identified some residues at the interface that influence binding. There are, however, no studies that have attempted to optimize the binding potential between MGP1 and TfR. In pursuits for finding therapeutic solutions for the New World arenaviruses, and to gain a greater understanding of MGP1 interactions with TfR, it is crucial to understand the structure-sequence relationship driving the interface formation. By displaying MGP1 on yeast surface we have examined the contributions of individual residues to the binding of solubilized ectodomain of TfR. We identified MGP1 binding hot spot residues, assessed the importance of posttranslational N-glycan modifications, and used a selection with random mutagenesis for affinity maturation. We show that the optimized MGP1 variants can bind more strongly to TfR than the native MGP1, and there is an MGP1 sequence that retains binding in the absence of glycosylation, but with the addition of further amino acid substitutions. The engineered variants can be used to probe cellular internalization or the blood-brain barrier crossing to achieve greater understanding of TfR mediated internalization.


Antigens, CD , Arenaviruses, New World/chemistry , Receptors, Transferrin , Viral Envelope Proteins , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/metabolism , Humans , Models, Molecular , Mutation , Protein Engineering , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
9.
Angew Chem Int Ed Engl ; 59(42): 18546-18555, 2020 10 12.
Article En | MEDLINE | ID: mdl-32627326

Tumor cell-surface markers are usually overexpressed or mutated protein receptors for which spatiotemporal regulation differs between and within cancers. Single-molecule fluorescence imaging can profile individual markers in different cellular contexts with molecular precision. However, standard single-molecule imaging methods based on overexpressed genetically encoded tags or cumbersome probes can significantly alter the native state of receptors. We introduce a live-cell points accumulation for imaging in nanoscale topography (PAINT) method that exploits aptamers as minimally invasive affinity probes. Localization and tracking of individual receptors are based on stochastic and transient binding between aptamers and their targets. We demonstrated single-molecule imaging of a model tumor marker (EGFR) on a panel of living cancer cells. Affinity to EGFR was finely tuned by rational engineering of aptamer sequences to define receptor motion and/or native receptor density.


Aptamers, Nucleotide/metabolism , Single Molecule Imaging/methods , Aptamers, Nucleotide/chemistry , Cell Line, Tumor , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Humans , Microscopy, Fluorescence , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism
10.
Proteins ; 88(12): 1569-1577, 2020 12.
Article En | MEDLINE | ID: mdl-32592192

Supply of iron into human cells is achieved by iron carrier protein transferrin and its receptor that upon complex formation get internalized by endocytosis. Similarly, the iron needs to be delivered into the brain, and necessitates the transport across the blood-brain barrier. While there are still unanswered questions about these mechanisms, extensive efforts have been made to use the system for delivery of therapeutics into biological compartments. The dimeric form of the receptor, where each subunit consists of three domains, further complicates the detailed investigation of molecular determinants responsible for guiding the receptor interactions with other proteins. Especially the apical domain's biological function has been elusive. To further the study of transferrin receptor, we have computationally decoupled the apical domain for soluble expression, and validated the design strategy by structure determination. Besides presenting a methodology for solubilizing domains, the results will allow for study of apical domain's function.


Computational Biology/methods , Protein Conformation , Protein Engineering , Protein Interaction Domains and Motifs , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Humans
11.
Pharm Dev Technol ; 25(9): 1063-1070, 2020 Nov.
Article En | MEDLINE | ID: mdl-32589499

In this work, T7 modified nanoliposome loaded SNF (T7-SNF-NLPs) was developed. The physicochemical properties and characteristics of T7-SNF-NLPs, including morphology, particle size, zeta potential, stability, and in vitro release, were determined. In vitro toxicity and cellular uptake were evaluated in RKO cells. Antitumor efficiency was examined in RKO cells-bearing Kunming mice to assess their potential applications in the development of nanoliposomes therapeutics. The average particle size of T7-SNF-NLPs was observed to be 131.6 ± 1.7 nm and the polydispersity index represented a uniform mono-dispersion with PDI = 0.19. SNF was sustainably released from T7-SNF-NLPs at a release rate of 65% at 48 h in pH 7.4 PBS. The release rate of SNF was over 72% from T7-SNF-NLPs in pH 6.5 PBS, faster than that in pH 7.4, which indicated that the release rate of SNF was enhanced under the acidic environment. In vitro study clearly showed that T7 modified NLPs was more effective in inducing uptake and apoptosis in cancer cells than nonmodified NLPs. The IC50 values of T7-SNF-NLPs treated RKO cells was 9.54 µg/mL, 9.23 µg/mL for SNF-NLPs and 16.85 µg/mL for free SNF. T7-SNF-NLPs was highly efficient in suppressing the tumor growth in xenograft tumor model. The proportion of Ki67 in T7-SNF-NLPs group was significantly lower than that of either free drug or nonmodified NLPs.


Antineoplastic Agents/chemistry , Collagen Type IV/chemistry , Colonic Neoplasms/drug therapy , Liposomes/chemistry , Nanoparticles/chemistry , Peptide Fragments/chemistry , Receptors, Transferrin/chemistry , Sorafenib/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Delivery Systems/methods , Humans , Mice , Particle Size , Sorafenib/pharmacology
12.
Nanotheranostics ; 4(3): 119-128, 2020.
Article En | MEDLINE | ID: mdl-32328439

Rational: Many efforts have been made to develop ligand-directed nanotheranostics in cancer management which could afford both therapeutic and diagnostic functions as well as tumor-tailored targeting. Theranostic nanoplatform targeting transferrin receptor (TfR) is an effective system for favorable delivery of diagnostic and therapeutic agents to malignancy site. Methods: To enable amalgamation of therapy and diagnosis to many TfR+ tumor, hTfR (human TfR) monoclonal antibody (mAb)-functionalized HPPS nanoparticle (HPPS-mAb) was prepared with hTfR mAb on the shell and with fluorophore DiR-BOA in the core. The targeting specificity was investigated in vitro by immunostaining and in vivo using a double-tumor-engrafted mouse model. HPPS-mAb/siRNA effect on HepG2 cells was determined by RT-PCR and western blot. Results: HPPS-mAb could specifically target cancer cells through TfR and achieve tumor accumulation at an early valuable time node, thus efficiently delivering therapeutic survivin siRNA into TfR+ HepG2 cells and mediating cell apoptosis. DiR-BOA can act as an imaging tool to diagnose cancer. Conclusions: Our studies provide a promising TfR mAb-directed theranostic nanoplatform candidate in tumor molecular imaging and in TfR targeted tumor therapy.


Antibodies, Monoclonal , Drug Delivery Systems/methods , Nanoparticles , Receptors, Transferrin , Theranostic Nanomedicine/methods , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Female , HeLa Cells , Hep G2 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/metabolism , Phospholipids/chemistry , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism
13.
Theranostics ; 10(10): 4308-4322, 2020.
Article En | MEDLINE | ID: mdl-32292496

Rationale: The dual-targeted drug delivery system was designed for enhancing permeation of the blood-brain barrier (BBB) and providing an anti-glioma effect. As transferrin receptor (TfR) is over-expressed by the brain capillary endothelial (hCMEC/D3) and glioma cells, a mouse monoclonal antibody, RI7217, with high affinity and selectivity for TfR, was used to study the brain targeted drug delivery system. Muscone, an ingredient of traditional Chinese medicine (TCM) musk, was used as the "guide" drug to probe the permeability of the BBB for drug delivery into the cerebrospinal fluid. This study investigated the combined effects of TCM aromatic resuscitation and modern receptor-targeted technology by the use of muscone/RI7217 co-modified docetaxel (DTX) liposomes for enhanced drug delivery to the brain for anti-glioma effect. Methods: Cellular drug uptake from the formulations was determined using fluorescence microscopy and flow cytometry. The drug penetrating ability into tumor spheroids were visualized using confocal laser scanning microscopy (CLSM). In vivo glioma-targeting ability of formulations was evaluated using whole-body fluorescent imaging system. The survival curve study was performed to evaluate the anti-glioma effect of the formulations. Results: The results showed that muscone and RI7217 co-modified DTX liposomes enhanced uptake into both hCMEC/D3 and U87-MG cells, increased penetration to the deep region of U87-MG tumor spheroids, improved brain targeting in vivo and prolonged survival time of nude mice bearing tumor. Conclusion: Muscone and RI7217 co-modified DTX liposomes were found to show improved brain targeting and enhanced the efficacy of anti-glioma drug treatment in vivo.


Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Cycloparaffins/pharmacology , Glioma/drug therapy , Liposomes/pharmacokinetics , Animals , Antigens, CD/chemistry , Antigens, CD/pharmacology , Blood-Brain Barrier/metabolism , Case-Control Studies , Cell Line, Tumor , Cell Survival/drug effects , Cycloparaffins/administration & dosage , Cycloparaffins/cerebrospinal fluid , Docetaxel/pharmacology , Drug Delivery Systems , Drug Therapy, Combination/methods , Glioma/metabolism , Liposomes/chemistry , Medicine, Chinese Traditional/adverse effects , Medicine, Chinese Traditional/methods , Mice , Mice, Nude , Permeability/drug effects , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Tubulin Modulators/pharmacology
14.
J Mol Biol ; 432(14): 3989-4009, 2020 06 26.
Article En | MEDLINE | ID: mdl-32304700

The impenetrability of the blood-brain barrier (BBB) to most conventional drugs impedes the treatment of central nervous system (CNS) disorders. Interventions for diseases like brain cancer, neurodegeneration, or age-associated inflammatory processes require varied approaches to CNS drug delivery. Cystine-dense peptides (CDPs) have drawn recent interest as drugs or drug-delivery vehicles. Found throughout the phylogenetic tree, often in drug-like roles, their size, stability, and protein interaction capabilities make CDPs an attractive mid-size biologic scaffold to complement conventional antibody-based drugs. Here, we describe the identification, maturation, characterization, and utilization of a CDP that binds to the transferrin receptor (TfR), a native receptor and BBB transporter for the iron chaperone transferrin. We developed variants with varying binding affinities (KD as low as 216 pM), co-crystallized it with the receptor, and confirmed murine cross-reactivity. It accumulates in the mouse CNS at ~25% of blood levels (CNS blood content is only ~1%-6%) and delivers neurotensin, an otherwise non-BBB-penetrant neuropeptide, at levels capable of modulating CREB signaling in the mouse brain. Our work highlights the utility of CDPs as a diverse, easy-to-screen scaffold family worthy of inclusion in modern drug discovery strategies, demonstrated by the discovery of a candidate CNS drug delivery vehicle ready for further optimization and preclinical development.


Blood-Brain Barrier/drug effects , Central Nervous System Diseases/drug therapy , Drug Delivery Systems , Peptides/pharmacology , Animals , Antigens, CD/chemistry , Antigens, CD/drug effects , Antigens, CD/genetics , Antigens, CD/pharmacology , Central Nervous System/drug effects , Cystine/chemistry , Cystine/genetics , Humans , Inflammation/drug therapy , Inflammation/pathology , Mice , Neuropeptides/chemistry , Neuropeptides/pharmacology , Neurotensin/chemistry , Neurotensin/pharmacology , Peptides/chemistry , Protein Binding/drug effects , Receptors, Transferrin/chemistry , Receptors, Transferrin/drug effects , Receptors, Transferrin/genetics
15.
J Biol Chem ; 295(7): 2068-2083, 2020 02 14.
Article En | MEDLINE | ID: mdl-31915245

Many secretory proteins are activated by cleavage at specific sites. The proprotein convertases (PCs) form a family of nine secretory subtilisin-like serine proteases, seven of which cleave at specific basic residues within the trans-Golgi network, granules, or at the cell surface/endosomes. The seventh member, PC7, is a type-I transmembrane (TM) protein with a 97-residue-long cytosolic tail (CT). PC7 sheds human transferrin receptor 1 (hTfR1) into soluble shTfR1 in endosomes. To better understand the physiological roles of PC7, here we focused on the relationship between the CT-regulated trafficking of PC7 and its ability to shed hTfR1. Deletion of the TMCT resulted in soluble PC7 and loss of its hTfR1 shedding activity. Extensive CT deletions and mutagenesis analyses helped us zoom in on three residues in the CT, namely Glu-719, Glu-721, and Leu-725, that are part of a novel motif, EXEXXXL725, critical for PC7 activity on hTfR1. NMR studies of two 14-mer peptides mimicking this region of the CT and its Ala variants revealed that the three exposed residues are on the same side of the molecule. This led to the identification of adaptor protein 2 (AP-2) as a protein that recognizes the EXEXXXL725 motif, thus representing a potentially new regulator of PC7 trafficking and cleavage activity. Immunocytochemistry of the subcellular localization of PC7 and its Ala variants of Leu-725 and Glu-719 and Glu-721 revealed that Leu-725 enhances PC7 localization to early endosomes and that, together with Glu-719 and Glu-721, it increases the endosomal activity of PC7 on hTfR1.


Antigens, CD/genetics , Cytosol/metabolism , Protein Transport/genetics , Receptors, Transferrin/genetics , Subtilisins/genetics , Transcription Factor AP-2/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence/genetics , Antigens, CD/chemistry , Cell Membrane/genetics , Cell Movement/genetics , Cytosol/chemistry , Endosomes/genetics , HEK293 Cells , Humans , Receptors, Transferrin/chemistry , Subtilisins/chemistry , trans-Golgi Network/genetics
16.
Nat Commun ; 11(1): 67, 2020 01 03.
Article En | MEDLINE | ID: mdl-31900422

Certain arenaviruses that circulate in rodent populations can cause life-threatening hemorrhagic fevers when they infect humans. Due to their efficient transmission, arenaviruses pose a severe risk for outbreaks and might be exploited as biological weapons. Effective countermeasures against these viruses are highly desired. Ideally, a single remedy would be effective against many or even all the pathogenic viruses in this family. However, despite the fact that all pathogenic arenaviruses from South America utilize transferrin receptor 1 (TfR1) as a cellular receptor, their viral glycoproteins are highly diversified, impeding efforts to isolate cross-neutralizing antibodies. Here we address this problem using a rational design approach to target TfR1-tropic arenaviruses with high potency and breadth. The pan-reactive molecule is highly effective against all arenaviruses that were tested, offering a universal therapeutic approach. Our design scheme avoids the shortcomings of previous immunoadhesins and can be used to combat other zoonotic pathogens.


Arenaviridae Infections/therapy , Arenavirus/immunology , Immunotherapy , Receptors, Transferrin/chemistry , Receptors, Transferrin/immunology , Receptors, Virus/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Arenavirus/chemistry , Arenavirus/genetics , Drug Design , Humans , Receptors, Transferrin/genetics , Receptors, Virus/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
17.
Nanoscale ; 12(1): 173-188, 2020 Jan 07.
Article En | MEDLINE | ID: mdl-31803890

Tumor necrosis factor (TNF-α) is capable of inducing apoptosis and is a promising candidate for genetic engineering drugs in cancer therapy; however, the serious side-effects of TNF-α hinder their clinical application. In the present study, a method for preparing fusion proteins of cell-penetrating peptides (CPP) and TNF-α (CTNF-α)-anchored exosomes coupled with superparamagnetic iron oxide nanoparticles (CTNF-α-exosome-SPIONs) with membrane targeting anticancer activity has been demonstrated. To acquire exosomes with TNF-α anchored in its membrane, a CTNF-α expression vector was constructed and a stable mesenchymal stem cell cell line that expressed CTNF-α was established. Conjugating transferrin-modified SPIONs (Tf-SPIONs) onto CTNF-α-exosomes through transferrin-transferrin receptor (Tf-TfR) interaction yields CTNF-α-exosome-SPIONs with good water dispersibility. The incorporation of TNF-α into exosomes and the conjugation of SPIONs significantly enhanced the binding capacity of TNF-α to its membrane-bound receptor TNFR I, thus increasing the therapeutic effects. CTNF-α-exosome-SPIONs significantly enhanced tumor cell growth inhibition via induction of the TNFR I-mediated apoptotic pathway. In vivo studies using murine melanoma subcutaneous cancer models showed that TNF-α-loaded exosome-based vehicle delivery enhanced cancer targeting under an external magnetic field and suppressed tumor growth with mitigating toxicity. Taken together, our results suggest that CTNF-α-exosome-SPIONs showed great potential in membrane targeting therapy.


Antineoplastic Agents/chemistry , Exosomes/metabolism , Magnetite Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Membrane/metabolism , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/metabolism , Exosomes/chemistry , Female , Ferric Compounds/chemistry , Half-Life , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Spectroscopy, Near-Infrared , Tissue Distribution , Transferrins/chemistry , Transferrins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Nat Microbiol ; 4(12): 2074-2081, 2019 12.
Article En | MEDLINE | ID: mdl-31636418

To maintain prolonged infection of mammals, African trypanosomes have evolved remarkable surface coats and a system of antigenic variation1. Within these coats are receptors for macromolecular nutrients such as transferrin2,3. These must be accessible to their ligands but must not confer susceptibility to immunoglobulin-mediated attack. Trypanosomes have a wide host range and their receptors must also bind ligands from diverse species. To understand how these requirements are achieved, in the context of transferrin uptake, we determined the structure of a Trypanosoma brucei transferrin receptor in complex with human transferrin, showing how this heterodimeric receptor presents a large asymmetric ligand-binding platform. The trypanosome genome contains a family of around 14 transferrin receptors4, which has been proposed to allow binding to transferrin from different mammalian hosts5,6. However, we find that a single receptor can bind transferrin from a broad range of mammals, indicating that receptor variation is unlikely to be necessary for promiscuity of host infection. In contrast, polymorphic sites and N-linked glycans are preferentially found in exposed positions on the receptor surface, not contacting transferrin, suggesting that transferrin receptor diversification is driven by a need for antigenic variation in the receptor to prolong survival in a host.


Host-Parasite Interactions/immunology , Immune Evasion , Receptors, Transferrin/chemistry , Receptors, Transferrin/immunology , Transferrin/metabolism , Trypanosoma brucei brucei/immunology , Antigenic Variation , Genetic Variation , Humans , Ligands , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Trypanosomiasis, African/immunology
19.
J Mater Chem B ; 7(38): 5814-5824, 2019 10 14.
Article En | MEDLINE | ID: mdl-31495855

Podophyllotoxin (PPT), a toxic polyphenol extracted from the roots of Podophyllum species, showed remarkable activity against P-glycoprotein (P-gp) mediated multidrug resistant (MDR) cancer cells. Many PPT-prodrugs based on nano-technology have been developed for increasing aqueous solubility and reducing the side effects of PPT; however, the sensitive linkers in almost all PPT-prodrugs were ester bonds, resulting in slow and incomplete drug release. We developed a redox/pH double-sensitive and tumor active targeted drug delivery system for PPT delivery, in which PPT was covalently coupled to T7-peptide (Pep) modified polyethylene glycol (PEG) or methoxy-polyethylene glycol (mPEG) through a disulfide bond to obtain the final polymer (Pep-PEG-SS-PPT or PEG-SS-PPT). The mixed micelles (Pep-SS-NPs) were made by mixing Pep-PEG-SS-PPT with PEG-SS-PPT, and the mixed micelles showed good size uniformity and high stability in serum solution. The in vitro release experiment showed that about (81.7 ± 2.8)% PPT was released from Pep-SS-NPs in 10 mM glutathione (GSH) at pH 7.4, and also about (64.6 ± 1.7)% PPT was released from Pep-SS-NPs at pH 5.0. In vitro cytotoxicity analysis suggested that Pep-SS-NPs exhibited 57- to 270-fold lower resistance index (RI) values for different drug-resistant cancer cell lines than paclitaxel (PTX) or docetaxel (DTX). The cell uptake assay indicated that the Pep-SS-NPs could significantly enhance the intracellular level of coumarin-6 compared to that of the control group. The maximum tolerated dose (MTD) of Pep-SS-NPs was increased greatly compared to that of free PPT (5.3-fold). In vivo research showed that Pep-SS-NPs significantly enhanced antitumor efficacy against MCF-7/ADR xenograft tumors compared to the control groups. These findings suggest that mixed micelles could be a potentially successful nanomedicine for MDR breast cancer therapy.


Drug Resistance, Multiple/drug effects , Micelles , Podophyllotoxin/chemistry , Prodrugs/pharmacology , Receptors, Transferrin/chemistry , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Survival/drug effects , Disulfides/chemistry , Female , Glutathione/chemistry , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Mice , Nanoparticles/chemistry , Oxidation-Reduction , Paclitaxel/pharmacology , Podophyllotoxin/metabolism , Podophyllotoxin/pharmacology , Polyethylene Glycols/chemistry , Prodrugs/chemistry , Prodrugs/therapeutic use , Receptors, Transferrin/metabolism
20.
Proc Natl Acad Sci U S A ; 116(41): 20462-20471, 2019 10 08.
Article En | MEDLINE | ID: mdl-31548398

Canine parvovirus (CPV) is an important pathogen causing severe diseases in dogs, including acute hemorrhagic enteritis, myocarditis, and cerebellar disease. Cross-species transmission of CPV occurs as a result of mutations on the viral capsid surface that alter the species-specific binding to the host receptor, transferrin receptor type-1 (TfR). The interaction between CPV and TfR has been extensively studied, and previous analyses have suggested that the CPV-TfR complex is asymmetric. To enhance the understanding of the underlying molecular mechanisms, we determined the CPV-TfR interaction using cryo-electron microscopy to solve the icosahedral (3.0-Å resolution) and asymmetric (5.0-Å resolution) complex structures. Structural analyses revealed conformational variations of the TfR molecules relative to the binding site, which translated into dynamic molecular interactions between CPV and TfR. The precise footprint of the receptor on the virus capsid was identified, along with the identity of the amino acid residues in the virus-receptor interface. Our "rock-and-roll" model provides an explanation for previous findings and gives insights into species jumping and the variation in host ranges associated with new pandemics in dogs.


Capsid/metabolism , Parvovirus, Canine/physiology , Receptors, Transferrin/metabolism , Receptors, Virus/metabolism , Virion/metabolism , Animals , Capsid/chemistry , Cats , Cryoelectron Microscopy , Dogs , Protein Conformation , Receptors, Transferrin/chemistry , Receptors, Virus/chemistry , Species Specificity , Virion/chemistry
...