Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Antioxid Redox Signal ; 38(1-3): 183-197, 2023 01.
Article in English | MEDLINE | ID: mdl-35754343

ABSTRACT

Aims: Though best known for its role in oxidative DNA damage repair, apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that regulates multiple host responses during oxidative stress, including the reductive activation of transcription factors. As knockout of the APE1-encoding gene, Apex1, is embryonically lethal, we sought to create a viable model with generalized inhibition of APE1 expression. Results: A hypomorphic (HM) mouse with decreased APE1 expression throughout the body was generated using a construct containing a neomycin resistance (NeoR) cassette knocked into the Apex1 site. Offspring were assessed for APE1 expression, breeding efficiency, and morphology with a focused examination of DNA damage in the stomach. Heterozygotic breeding pairs yielded 50% fewer HM mice than predicted by Mendelian genetics. APE1 expression was reduced up to 90% in the lungs, heart, stomach, and spleen. The HM offspring were typically smaller, and most had a malformed tail. Oxidative DNA damage was increased spontaneously in the stomachs of HM mice. Further, all changes were reversed when the NeoR cassette was removed. Primary gastric epithelial cells from HM mice differentiated more quickly and had more evidence of oxidative DNA damage after stimulation with Helicobacter pylori or a chemical carcinogen than control lines from wildtype mice. Innovation: A HM mouse with decreased APE1 expression throughout the body was generated and extensively characterized. Conclusion: The results suggest that HM mice enable studies of APE1's multiple functions throughout the body. The detailed characterization of the stomach showed that gastric epithelial cells from HM were more susceptible to DNA damage. Antioxid. Redox Signal. 38, 183-197.


Subject(s)
DNA Repair , Oxidative Stress , Mice , Animals , DNA Damage , Oxidation-Reduction , Disease Models, Animal , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Stomach , Endonucleases/genetics , Endonucleases/metabolism
2.
Front Immunol ; 11: 553994, 2020.
Article in English | MEDLINE | ID: mdl-33603730

ABSTRACT

Pathogenic intestinal bacteria lead to significant disease in humans. Here we investigated the role of the multifunctional protein, Apurinic/apyrimidinic endonuclease 1 (APE1), in regulating the internalization of bacteria into the intestinal epithelium. Intestinal tumor-cell lines and primary human epithelial cells were infected with Salmonella enterica serovar Typhimurium or adherent-invasive Escherichia coli. The effects of APE1 inhibition on bacterial internalization, the regulation of Rho GTPase Rac1 as well as the epithelial cell barrier function were assessed. Increased numbers of bacteria were present in APE1-deficient colonic tumor cell lines and primary epithelial cells. Activation of Rac1 was augmented following infection but negatively regulated by APE1. Pharmacological inhibition of Rac1 reversed the increase in intracellular bacteria in APE1-deficient cells whereas overexpression of constitutively active Rac1 augmented the numbers in APE1-competent cells. Enhanced numbers of intracellular bacteria resulted in the loss of barrier function and a delay in its recovery. Our data demonstrate that APE1 inhibits the internalization of invasive bacteria into human intestinal epithelial cells through its ability to negatively regulate Rac1. This activity also protects epithelial cell barrier function.


Subject(s)
Colon , DNA-(Apurinic or Apyrimidinic Site) Lyase/immunology , Epithelial Cells , Escherichia coli Infections , Escherichia coli/immunology , Intestinal Mucosa , Salmonella Infections , Salmonella typhimurium/immunology , rac1 GTP-Binding Protein/immunology , Colon/immunology , Colon/microbiology , Colon/pathology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , HT29 Cells , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Salmonella Infections/immunology , Salmonella Infections/pathology
3.
PLoS Pathog ; 12(1): e1005382, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26761793

ABSTRACT

Generation of reactive oxygen species (ROS) during infection is an immediate host defense leading to microbial killing. APE1 is a multifunctional protein induced by ROS and after induction, protects against ROS-mediated DNA damage. Rac1 and NAPDH oxidase (Nox1) are important contributors of ROS generation following infection and associated with gastrointestinal epithelial injury. The purpose of this study was to determine if APE1 regulates the function of Rac1 and Nox1 during oxidative stress. Gastric or colonic epithelial cells (wild-type or with suppressed APE1) were infected with Helicobacter pylori or Salmonella enterica and assessed for Rac1 and NADPH oxidase-dependent superoxide production. Rac1 and APE1 interactions were measured by co-immunoprecipitation, confocal microscopy and proximity ligation assay (PLA) in cell lines or in biopsy specimens. Significantly greater levels of ROS were produced by APE1-deficient human gastric and colonic cell lines and primary gastric epithelial cells compared to control cells after infection with either gastric or enteric pathogens. H. pylori activated Rac1 and Nox1 in all cell types, but activation was higher in APE1 suppressed cells. APE1 overexpression decreased H. pylori-induced ROS generation, Rac1 activation, and Nox1 expression. We determined that the effects of APE1 were mediated through its N-terminal lysine residues interacting with Rac1, leading to inhibition of Nox1 expression and ROS generation. APE1 is a negative regulator of oxidative stress in the gastrointestinal epithelium during bacterial infection by modulating Rac1 and Nox1. Our results implicate APE1 in novel molecular interactions that regulate early stress responses elicited by microbial infections.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Intestinal Mucosa/immunology , Salmonella Infections/immunology , rac1 GTP-Binding Protein/metabolism , Blotting, Western , Cell Line , Fluorescent Antibody Technique , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter Infections/metabolism , Humans , Immunoprecipitation , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Microscopy, Confocal , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Salmonella Infections/metabolism , rac1 GTP-Binding Protein/immunology
4.
Cell Rep ; 8(5): 1558-70, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25176655

ABSTRACT

Tumor cell extravasation is a key step during cancer metastasis, yet the precise mechanisms that regulate this dynamic process are unclear. We utilized a high-resolution time-lapse intravital imaging approach to visualize the dynamics of cancer cell extravasation in vivo. During intravascular migration, cancer cells form protrusive structures identified as invadopodia by their enrichment of MT1-MMP, cortactin, Tks4, and importantly Tks5, which localizes exclusively to invadopodia. Cancer cells extend invadopodia through the endothelium into the extravascular stroma prior to their extravasation at endothelial junctions. Genetic or pharmacological inhibition of invadopodia initiation (cortactin), maturation (Tks5), or function (Tks4) resulted in an abrogation of cancer cell extravasation and metastatic colony formation in an experimental mouse lung metastasis model. This provides direct evidence of a functional role for invadopodia during cancer cell extravasation and distant metastasis and reveals an opportunity for therapeutic intervention in this clinically important process.


Subject(s)
Cell Surface Extensions/metabolism , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Transcellular Cell Migration , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Cell Line, Tumor , Cell Surface Extensions/drug effects , Chick Embryo , Cortactin/genetics , Cortactin/metabolism , Female , Humans , Lung Neoplasms/pathology , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Nude , Neoplasm Metastasis , Neoplastic Stem Cells/physiology , Phosphate-Binding Proteins , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology
5.
Mol Pharm ; 10(1): 33-42, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-22731633

ABSTRACT

Nanomaterials with elongated architectures have been shown to possess differential tumor homing properties compared to their spherical counterparts. Here, we investigate whether this phenomenon is mirrored by plant viral nanoparticles that are filamentous (Potato virus X) or spherical (Cowpea mosaic virus). Our studies demonstrate that Potato virus X (PVX) and Cowpea mosaic virus (CPMV) show distinct biodistribution profiles and differ in their tumor homing and penetration efficiency. Analogous to what is seen with inorganic nanomaterials, PVX shows enhanced tumor homing and tissue penetration. Human tumor xenografts exhibit higher uptake of PEGylated filamentous PVX compared to CPMV, particularly in the core of the tumor. This is supported by immunohistochemical analysis of the tumor sections, which indicates greater penetration and accumulation of PVX within the tumor tissues. The enhanced tumor homing and retention properties of PVX along with its higher payload carrying capacity make it a potentially superior platform for applications in cancer drug delivery and imaging applications.


Subject(s)
Comovirus/metabolism , Nanoparticles/administration & dosage , Neoplasms/drug therapy , Neoplasms/virology , Potexvirus/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Chick Embryo , Diagnostic Imaging/methods , Drug Delivery Systems/methods , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/pathology , Oncolytic Virotherapy/methods , Plants/virology , Polyethylene Glycols/administration & dosage , Tissue Distribution
6.
Cancer Res ; 72(15): 3851-63, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22855743

ABSTRACT

Tumor vascularization is requisite for breast cancer progression, and high microvascular density in tumors is a poor prognostic indicator. Patients bearing breast cancers expressing human embryonic stem cell (hESC)-associated genes similarly exhibit high mortality rates, and the expression of embryonic proteins is associated with tumor progression. Here, we show that Nodal, a hESC-associated protein, promotes breast cancer vascularization. We show that high levels of Nodal are positively correlated with high vascular densities in human breast lesions (P = 0.0078). In vitro, we show that Nodal facilitates breast cancer-induced endothelial cell migration and tube formation, largely by upregulating the expression and secretion of proangiogenic factors by breast cancer cells. Using a directed in vivo angiogenesis assay and a chick chorioallantoic membrane assay, we show that Nodal promotes vascular recruitment in vivo. In a clinically relevant in vivo model, whereby Nodal expression was inhibited following tumor formation, we found a significant reduction in tumor vascularization concomitant with elevated hypoxia and tumor necrosis. These findings establish Nodal as a potential target for the treatment of breast cancer angiogenesis and progression.


Subject(s)
Breast Neoplasms/blood supply , Carcinoma/blood supply , Neovascularization, Pathologic/genetics , Nodal Protein/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Cell Count , Cells, Cultured , Chick Embryo , Disease Progression , Embryo, Mammalian/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice , Mice, Nude , Microvessels/pathology , Neovascularization, Pathologic/pathology , Nodal Protein/genetics , Nodal Protein/metabolism , Stem Cell Niche/genetics , Transfection
7.
J Biol Chem ; 287(34): 29184-93, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22753409

ABSTRACT

Pannexin 1 (Panx1) is a channel-forming glycoprotein expressed in different cell types of mammalian skin. We examined the role of Panx1 in melanoma tumorigenesis and metastasis since qPCR and Western blots revealed that mouse melanocytes exhibited low levels of Panx1 while increased Panx1 expression was correlated with tumor cell aggressiveness in the isogenic melanoma cell lines (B16-F0, -F10, and -BL6). Panx1 shRNA knockdown (Panx1-KD) generated stable BL6 cell lines, with reduced dye uptake, that showed a marked increase in melanocyte-like cell characteristics including higher melanin production, decreased cell migration and enhanced formation of cellular projections. Western blotting and proteomic analyses using 2D-gel/mass spectroscopy identified vimentin and ß-catenin as two of the markers of malignant melanoma that were down-regulated in Panx1-KD cells. Xenograft Panx1-KD cells grown within the chorioallantoic membrane of avian embryos developed tumors that were significantly smaller than controls. Mouse-Alu qPCR of the excised avian embryonic organs revealed that tumor metastasis to the liver was significantly reduced upon Panx1 knockdown. These data suggest that while Panx1 is present in skin melanocytes it is up-regulated during melanoma tumor progression, and tumorigenesis can be inhibited by the knockdown of Panx1 raising the possibility that Panx1 may be a viable target for the treatment of melanoma.


Subject(s)
Connexins/biosynthesis , Gene Expression Regulation, Neoplastic , Melanins/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/biosynthesis , Animals , Cell Line, Tumor , Cell Movement/genetics , Connexins/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Melanins/genetics , Melanocytes/pathology , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Up-Regulation/genetics , Vimentin/genetics , Vimentin/metabolism , beta Catenin/genetics , beta Catenin/metabolism
8.
PLoS One ; 7(1): e30177, 2012.
Article in English | MEDLINE | ID: mdl-22276156

ABSTRACT

The analysis of dynamic events in the tumor microenvironment during cancer progression is limited by the complexity of current in vivo imaging models. This is coupled with an inability to rapidly modulate and visualize protein activity in real time and to understand the consequence of these perturbations in vivo. We developed an intravital imaging approach that allows the rapid induction and subsequent depletion of target protein levels within human cancer xenografts while assessing the impact on cell behavior and morphology in real time. A conditionally stabilized fluorescent E-cadherin chimera was expressed in metastatic breast cancer cells, and the impact of E-cadherin induction and depletion was visualized using real-time confocal microscopy in a xenograft avian embryo model. We demonstrate the assessment of protein localization, cell morphology and migration in cells undergoing epithelial-mesenchymal and mesenchymal-epithelial transitions in breast tumors. This technique allows for precise control over protein activity in vivo while permitting the temporal analysis of dynamic biophysical parameters.


Subject(s)
Breast Neoplasms/metabolism , Cadherins/metabolism , Animals , Birds/embryology , Cell Line, Tumor , Diagnostic Imaging , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Morpholines/pharmacokinetics , Morpholines/pharmacology , Transplantation, Heterologous , Vimentin/metabolism
9.
J Vis Exp ; (52)2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21730939

ABSTRACT

Current technologies for tumor imaging, such as ultrasound, MRI, PET and CT, are unable to yield high-resolution images for the assessment of nanoparticle uptake in tumors at the microscopic level(1,2,3,) highlighting the utility of a suitable xenograft model in which to perform detailed uptake analyses. Here, we use high-resolution intravital imaging to evaluate nanoparticle uptake in human tumor xenografts in a modified, shell-less chicken embryo model. The chicken embryo model is particularly well-suited for these in vivo analyses because it supports the growth of human tumors, is relatively inexpensive and does not require anesthetization or surgery 4,5. Tumor cells form fully vascularized xenografts within 7 days when implanted into the chorioallantoic membrane (CAM)( 6). The resulting tumors are visualized by non-invasive real-time, high-resolution imaging that can be maintained for up to 72 hours with little impact on either the host or tumor systems. Nanoparticles with a wide range of sizes and formulations administered distal to the tumor can be visualized and quantified as they flow through the bloodstream, extravasate from leaky tumor vasculature, and accumulate at the tumor site. We describe here the analysis of nanoparticles derived from Cowpea mosaic virus (CPMV) decorated with near-infrared fluorescent dyes and/or polyethylene glycol polymers (PEG) (7, 8, 9,10,11). Upon intravenous administration, these viral nanoparticles are rapidly internalized by endothelial cells, resulting in global labeling of the vasculature both outside and within the tumor(7,12). PEGylation of the viral nanoparticles increases their plasma half-life, extends their time in the circulation, and ultimately enhances their accumulation in tumors via the enhanced permeability and retention (EPR) effect (7, 10,11). The rate and extent of accumulation of nanoparticles in a tumor is measured over time using image analysis software. This technique provides a method to both visualize and quantify nanoparticle dynamics in human tumors.


Subject(s)
Colonic Neoplasms/metabolism , Nanoparticles/administration & dosage , Animals , Chick Embryo , Chorioallantoic Membrane/blood supply , Colonic Neoplasms/blood supply , Comovirus/chemistry , Fluorescent Dyes/chemistry , HT29 Cells , Humans , Image Processing, Computer-Assisted/methods , Microinjections/instrumentation , Microinjections/methods , Microscopy/methods , Nanoparticles/chemistry , Polyethylene Glycols/chemistry
10.
Small ; 7(12): 1664-72, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21520408

ABSTRACT

Multivalent nanoparticles have several key advantages in terms of solubility, binding avidity, and uptake, making them particularly well suited to molecular imaging applications. Herein is reported the stepwise synthesis and characterization of NIR viral nanoparticles targeted to gastrin-releasing peptide receptors that are over-expressed in human prostate cancers. The pan-bombesin analogue, [ß-Ala11, Phe13, Nle14]bombesin-(7-14), is conjugated to cowpea mosaic virus particles functionalized with an NIR dye (Alexa Fluor 647) and polyethylene glycol (PEG) using the copper(I)-catalyzed azide-alkyne cycloaddition reaction. Targeting and uptake in human PC-3 prostate cells is demonstrated in vitro. Tumor homing is observed using human prostate tumor xenografts on the chicken chorioallantoic membrane model using intravital imaging. Further development of this viral nanoparticle platform may open the door to potential clinical noninvasive molecular imaging strategies.


Subject(s)
Nanoparticles/chemistry , Prostatic Neoplasms/pathology , Receptors, Bombesin/metabolism , Animals , Bombesin/chemistry , Cell Line, Tumor , Chickens , Comovirus/chemistry , Humans , Male , Polyethylene Glycols/chemistry
11.
Lab Invest ; 91(8): 1181-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21502940

ABSTRACT

Maspin (mammary serine protease inhibitor or SerpinB5) acts as a tumor suppressor when overexpressed in aggressive cancer cell lines. However, its role in human cancer is controversial. Maspin expression has been associated with a poor prognosis in some studies, whereas in others, with favorable outcome. The clinical data suggest, however, that nuclear-localized maspin is associated with improved survival. We hypothesized that the tumor suppressor activity of maspin may require nuclear localization, and that the discordance between clinical and experimental reports is a consequence of the variable subcellular distribution of maspin. Furthermore, we surmized that nuclear maspin could function as a tumor suppressor through the regulation of genes involved in tumor growth and invasion. Maspin or maspin fused to a nuclear export signal were expressed in metastatic human breast and epidermoid carcinoma cell lines. We found that pan-cellular localized maspin inhibited in vivo tumor growth and metastasis when assessed in xenograft chicken embryo and murine mammary fat pad injection models. However, when maspin was excluded from the nucleus via a nuclear exclusion signal, it no longer functioned as a metastasis suppressor. Using chromatin immunoprecipitation, we show that nuclear maspin was enriched at the promoter of colony-stimulating factor-1 (CSF-1) and associated with diminished levels of CSF-1 mRNA. Our findings demonstrate that the nuclear localization of maspin is required for its tumor and metastasis suppressor functions in vivo, and suggest that its mechanism of action involves, in part, direct association of maspin with target genes.


Subject(s)
Cell Nucleus/metabolism , Neoplasm Metastasis , Serpins/metabolism , Animals , Breast Neoplasms/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Chromatin/metabolism , Female , Humans , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Nude , Promoter Regions, Genetic
12.
Nanomedicine (Lond) ; 6(2): 351-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21385137

ABSTRACT

AIMS: Vimentin, a type III intermediate filament, is upregulated during epithelial-mesenchymal transition and tumor progression. Vimentin is surface-expressed on cells involved in inflammation; the function remains unknown. We investigated the expression of surface vimentin on cancer cells and evaluated targeting nanoparticles to tumors exploiting vimentin. MATERIALS & METHODS: Cowpea mosaic virus nanoparticles that interact with surface vimentin were used as probes. Tumor homing was tested using the chick chorioallantoic membrane model with human tumor xenografts. RESULTS & DISCUSSION: Surface vimentin levels varied during cell cycle and among the cell lines tested. Surface vimentin expression correlated with cowpea mosaic virus uptake, underscoring the utility of cowpea mosaic virus to detect invasive cancer cells. Targeting to tumor xenografts was observed; homing was based on the enhanced permeability and retention effect. Our data provide novel insights into the role of surface vimentin in cancer and targeting nanoparticles in vivo.


Subject(s)
Cell Membrane Permeability , Comovirus/metabolism , Nanoparticles/ultrastructure , Neoplasms/metabolism , Vimentin/metabolism , Virion/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Chick Embryo , Comovirus/ultrastructure , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Vimentin/genetics , Virion/ultrastructure
13.
Nat Protoc ; 5(8): 1406-17, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20671724

ABSTRACT

Viral nanoparticles are a novel class of biomolecular agents that take advantage of the natural circulatory and targeting properties of viruses to allow the development of therapeutics, vaccines and imaging tools. We have developed a multivalent nanoparticle platform based on the cowpea mosaic virus (CPMV) that facilitates particle labeling at high density with fluorescent dyes and other functional groups. Compared with other technologies, CPMV-based viral nanoparticles are particularly suited for long-term intravital vascular imaging because of their biocompatibility and retention in the endothelium with minimal side effects. The stable, long-term labeling of the endothelium allows the identification of vasculature undergoing active remodeling in real time. In this study, we describe the synthesis, purification and fluorescent labeling of CPMV nanoparticles, along with their use for imaging of vascular structure and for intravital vascular mapping in developmental and tumor angiogenesis models. Dye-labeled viral nanoparticles can be synthesized and purified in a single day, and imaging studies can be conducted over hours, days or weeks, depending on the application.


Subject(s)
Comovirus/isolation & purification , Embryo, Mammalian/blood supply , Embryo, Nonmammalian/blood supply , Endothelium, Vascular/anatomy & histology , Nanoparticles , Nanotechnology/methods , Neoplasms/blood supply , Animals , Chickens , Embryo Culture Techniques , Fabaceae/virology , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Mice , Microinjections/methods , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods
14.
J Nanopart Res ; 12(5): 1599-1608, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-22328862

ABSTRACT

The imaging of molecular markers associated with disease offers the possibility for earlier detection and improved treatment monitoring. Receptors for gastrin-releasing peptide are overexpressed on prostate cancer cells offering a promising imaging target, and analogs of bombesin, an amphibian tetradecapeptide have been previously demonstrated to target these receptors. Therefore, the pan-bombesin analog [ß-Ala11, Phe13, Nle14]bombesin-(7-14) was conjugated through a linker to dye-functionalized superparamagnetic iron oxide nanoparticles for the development of a new potential magnetic resonance imaging probe. The peptide was conjugated via click chemistry, demonstrating a complementary alternative methodology to conventional peptide-nanoparticle conjugation strategies. The peptide-functionalized nanoparticles were then demonstrated to be selectively taken up by PC-3 prostate cancer cells relative to unfunctionalized nanoparticles and this uptake was inhibited by the presence of free peptide, confirming the specificity of the interaction. This study suggests that these nanoparticles have the potential to serve as magnetic resonance imaging probes for the detection of prostate cancer.

SELECTION OF CITATIONS
SEARCH DETAIL
...