Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Apoptosis ; 26(3-4): 209-218, 2021 04.
Article in English | MEDLINE | ID: mdl-33655467

ABSTRACT

Apoptosis plays an essential role in the pathophysiologic processes of rheumatoid arthritis. A molecular probe that allows spatiotemporal observation of apoptosis in vitro, in vivo, and ex vivo concomitantly would be useful to monitoring or predicting pathophysiologic stages. In this study we investigated whether cyclic apoptosis-targeting peptide-1 (CApoPep-1) can be used as an apoptosis imaging probe in inflammatory arthritis. We tested the utility of CApoPep-1 for detecting apoptotic immune cells in vitro and ex vivo using flow cytometry and immunofluorescence. The feasibility of visualizing and quantifying apoptosis using this probe was evaluated in a murine collagen-induced arthritis (CIA) model, especially after treatment. CApoPep-1 peptide may successfully replace Annexin V for in vitro and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay for ex vivo in the measurement of apoptotic cells, thus function as a sensitive probe enough to be used clinically. In vivo imaging in CIA mice revealed that CApoPep-1 had 42.9 times higher fluorescence intensity than Annexin V for apoptosis quantification. Furthermore, it may be used as an imaging probe for early detection of apoptotic response in situ after treatment. The CApoPep-1 signal was mostly co-localized with the TUNEL signal (69.6% of TUNEL+ cells) in defined cell populations in joint tissues of CIA mice. These results demonstrate that CApoPep-1 is sufficiently sensitive to be used as an apoptosis imaging probe for multipurpose applications which could detect the same target across in vitro, in vivo, to ex vivo in inflammatory arthritis.


Subject(s)
Arthritis/diagnostic imaging , Diagnostic Imaging/methods , Fluorescent Dyes/chemistry , Oligopeptides/chemistry , Animals , Apoptosis , Arthritis, Experimental/diagnostic imaging , Arthritis, Rheumatoid/diagnostic imaging , Disease Models, Animal , In Situ Nick-End Labeling/methods , Mice
2.
Biomacromolecules ; 17(1): 12-9, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26646195

ABSTRACT

Ferritin cage nanoparticles are promising platforms for targeted delivery of imaging and therapeutic agents. One of the main advantages of cage nanoparticles is the ability to display multiple functionalities through genetic modification so as to achieve desired therapeutic or diagnostic functions. Ferritin complexes formed from short ferritin (sFt) lacking the fifth helix can accommodate dual peptide and protein functionalities on N- and C-terminal sites in sFt monomers. The resulting double-chambered NanoCage (DCNC) offers the potential of dual activities; these activities are augmented by the avidity of the ligands, which do not impede each other's function. Here we demonstrated proof-of-concept of DCNCs, loading the tumor-targeting proapoptotic peptide CGKRK(KLAKLAK)2 onto the N-terminal chamber and green fluorescent protein (GFP) onto the C-terminal chamber. The resulting KLAK-sFt-GFP DCNCs were internalized into the human breast adenocarcinoma cell line MDA-MB-231 and induced apoptosis. These findings suggest that DCNCs containing various combinations of peptides and proteins could be applied as therapeutics in different diseases.


Subject(s)
Ferritins/chemistry , Green Fluorescent Proteins/chemistry , Peptides/chemistry , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Humans , Ligands , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanotechnology/methods
3.
PLoS One ; 10(2): e0118310, 2015.
Article in English | MEDLINE | ID: mdl-25705892

ABSTRACT

We developed and tested a multicomponent peptide-woven siRNA nanocomplex (PwSN) comprising different peptides designed for efficient cellular targeting, endosomal escape, and release of siRNA. To enhance tumor-specific cellular uptake, we connected an interleukin-4 receptor-targeting peptide (I4R) to a nine-arginine peptide (9r), yielding I4R-9r. To facilitate endosomal escape, we blended endosomolytic peptides into the I4R-9r to form a multicomponent nanocomplex. Lastly, we modified 9r peptides by varying the number and positions of positive charges to obtain efficient release of siRNA from the nanocomplex in the cytosol. Using this step-wise approach for overcoming the biological challenges of siRNA delivery, we obtained an optimized PwSN with significant biological activity in vitro and in vivo. Interestingly, surface plasmon resonance analyses and three-dimensional peptide models demonstrated that our designed peptide adopted a unique structure that was correlated with faster complex disassembly and a better gene-silencing effect. These studies further elucidate the siRNA nanocomplex delivery pathway and demonstrate the applicability of our stepwise strategy to the design of siRNA carriers capable of overcoming multiple challenges and achieving efficient delivery.


Subject(s)
Drug Delivery Systems/methods , Nanoparticles/chemistry , Peptides/chemistry , RNA, Small Interfering/administration & dosage , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Amino Acid Sequence , Animals , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HT29 Cells , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Nanoparticles/administration & dosage , Peptides/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Receptors, Interleukin-4/chemistry , Receptors, Interleukin-4/genetics , Reproducibility of Results , Surface Plasmon Resonance , Transplantation, Heterologous
4.
ACS Nano ; 7(9): 7462-71, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23927443

ABSTRACT

Protein-cage nanoparticles are promising multifunctional platforms for targeted delivery of imaging and therapeutic agents owing to their biocompatibility, biodegradability, and low toxicity. The major advantage of protein-cage nanoparticles is the ability to decorate their surfaces with multiple functionalities through genetic and chemical modification to achieve desired properties for therapeutic and/or diagnostic purposes. Specific peptides identified by phage display can be genetically fused onto the surface of cage proteins to promote the association of nanoparticles with a particular cell type or tissue. Upon symmetrical assembly of the cage, peptides are clustered on the surface of the cage protein in bunches. The resulting PBNC (peptide bunches on nanocage) offers the potential of synergistically increasing the avidity of the peptide ligands, thereby enhancing their blocking ability for therapeutic purposes. Here, we demonstrated a proof-of-principle of PBNCs, fusing the interleukin-4 receptor (IL-4R)-targeting peptide, AP-1, identified previously by phage display, with ferritin-L-chain (FTL), which undergoes 24-subunit assembly to form highly stable AP-1-containing nanocage proteins (AP1-PBNCs). AP1-PBNCs bound specifically to the IL-4R-expressing cell line, A549, and their binding and internalization were specifically blocked by anti-IL-4R antibody. AP1-PBNCs exhibited dramatically enhanced binding avidity to IL-4R compared with AP-1 peptide, measured by surface plasmon resonance spectroscopy. Furthermore, treatment with AP1-PBNCs in a murine model of experimental asthma diminished airway hyper-responsiveness and eosinophilic airway inflammation along with decreased mucus hyperproduction. These findings hold great promise for the application of various PBNCs with ligand-specific peptides in therapeutics for different diseases, such as cancer.


Subject(s)
Asthma/drug therapy , Asthma/immunology , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Peptides/administration & dosage , Peptides/immunology , Receptors, Interleukin-4/immunology , Animals , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/chemical synthesis , Anti-Asthmatic Agents/immunology , Cell Line, Tumor , Drug Design , Female , Humans , Mice , Mice, Inbred C57BL , Nanocapsules/ultrastructure , Peptides/chemistry , Protein Binding , Treatment Outcome
5.
J Cell Mol Med ; 12(5B): 2003-14, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19012727

ABSTRACT

Imaging or drug delivery tools for atherosclerosis based on the plaque biology are still insufficient. Here, we attempted to identify peptides that selectively home to atherosclerotic plaques using phage display. A phage library containing random peptides was ex vivo screened for binding to human atheroma tissues. After three to four rounds of selection, the DNA inserts of phage clones wer sequenced. A peptide sequence, CRKRLDRNC, was the most frequently occurring one. Intravenously injected phage displaying the CRKRLDRNC peptide was observed to home to atherosclerotic aortic tissues of low-density lipoprotein receptor-deficient (Ldlr-/-) mice at higher levels than to normal aortic tissues of wild-type mice. Moreover, a fluorescein- or radioisotope-conjugated synthetic CRKRLDRNC peptide, but not a control peptide, homed in vivo to atherosclerotic plaques in Ldlr(-/-) mice, while homing of the peptide to other organs such as brain was minimal. The homing peptide co-localized with endothelial cells, macrophages and smooth muscle cells a mouse and human atherosclerotic plaques. Homology search revealed that the CRKRLDRNC peptide shares a motif of interleukin-receptor (IL-4) that is critical for binding to its receptor. The peptide indeed co-localized with IL-4 receptor (IL-4R) at atherosclerotic plaques. Moreover, the peptide bound to cultured cells expressing IL-4R on the cell surface and the binding was inhibited by the knock-down of IL-4R. These results show that the CRKRLDRNC peptide homes to atherosclerotic plaques through binding to IL-4R as its target and may be a useful tool for selective drug delivery and molecular imaging of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Library , Receptors, Interleukin-4/metabolism , Adult , Aged , Amino Acid Sequence , Animals , Atherosclerosis/genetics , Autoradiography , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Oligopeptides/genetics , Protein Binding/genetics , Receptors, Interleukin-4/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism
6.
J Nucl Med ; 49(9): 1480-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18703598

ABSTRACT

UNLABELLED: We investigated the feasibility of using combination gene therapy and noninvasive nuclear imaging after expression of the human sodium iodide symporter (hNIS) and inhibition of the multidrug resistance (MDR1) gene in colon cancer cells. METHODS: HCT-15 cells were stably transfected with a dual expression vector, in which the hNIS gene, driven by a constitutive cytomegalovirus promoter, has been coupled to an MDR1 short hairpin RNA (shRNA) cassette. Cell lines stably expressing the hNIS gene and MDR1 shRNA (designated MN-61 and MN-62) were produced, and the expression of the NIS gene and MDR1 shRNA was examined by Western blotting, reverse transcription-polymerase chain reaction, and immunostaining. The functional activities of MDR1 shRNA were determined by paclitaxel uptake and sensitivity to doxorubicin. Functional NIS expression was confirmed by the uptake and efflux of (125)I and the cytotoxicity of (131)I. The effect of the combination of (131)I and doxorubicin was determined by an in vitro clonogenic assay. In vivo NIS expression was examined by small-animal PET with (124)I. RESULTS: The shMDR-NIS-expressing cells showed a significant decrease in the expression of MDR1 messenger RNA and its translated product, P-glycoprotein. The inhibition of P-glycoprotein expression by shRNA enhanced the intracellular accumulation of paclitaxel, the cellular retention of which is mediated by P-glycoprotein, thereby increasing sensitivity to the anticancer drug. The shMDR-NIS-expressing cells showed a significant increase of (125)I uptake, which was completely inhibited by KClO(4). Although the iodide efflux rate was rapid, the cell survival rate was markedly reduced by (131)I treatment. Interestingly, the combination of doxorubicin and a radioiodide ((131)I) displayed synergistic cytotoxicity that correlated with MDR1 inhibition and NIS expression in shMDR-NIS-expressing cells. Furthermore, in mice with shMDR-NIS-expressing tumor xenografts, small-animal PET with (124)I clearly visualized shMDR1-NIS-expressing tumors. CONCLUSION: We developed a dual expression vector with the NIS gene and MDR1 shRNA. This study represents a promising first step in investigations of the potential use of a combination of the NIS gene and MDR1 shRNA as a new therapeutic strategy allowing RNA interference-based gene therapy, NIS-based radioiodine therapy, and in vivo monitoring based on NIS imaging.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Colonic Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/genetics , RNA/genetics , RNA/therapeutic use , Symporters/genetics , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Combined Modality Therapy , Mice , Transfection/methods , Treatment Outcome
7.
FEBS Lett ; 582(15): 2161-6, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18519035

ABSTRACT

Stabilin-2 was recently identified as a novel receptor for membrane phosphatidylserine of apoptotic cells. To identify proteins that were candidates for stabilin-2 cytoplasmic domain binding, we screened a human spleen cDNA library using the yeast two-hybrid system. We found that thymosin beta4 interacts with the stabilin-2 cytoplasmic domain and is co-localized with stabilin-2 at the phagocytic cup. Knockdown of thymosin beta4 significantly decreased the phagocytic activity of stabilin-2, whereas overexpression of thymosin beta4 increased this activity. Additionally, amino acids 2504-2514 of stabilin-2 cytoplasmic domain were found to be responsible for the interaction with thymosin beta4. Taken together, these results suggest that thymosin beta4 is a downstream molecule of stabilin-2 that plays a role in stabilin-2-mediated cell corpse clearance.


Subject(s)
Apoptosis , Cell Adhesion Molecules, Neuronal/metabolism , Phagocytosis , Thymosin/metabolism , Amino Acid Sequence , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Gene Library , Humans , Molecular Sequence Data , Thymosin/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...