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










Publication year range
1.
Nanoscale ; 16(7): 3729-3737, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38294340

ABSTRACT

Lanthanide-based macrocycles are successfully incorporated into hybrid polyionic complexes, formed by adding a mixture of zirconium ions to a solution of a double-hydrophilic block copolymer. The resulting nanoobjects with an average radius of approximately 10-15 nm present good colloidal and chemical stability in physiological media even in the presence of competing ions such as phosphate or calcium ions. The final optical and magnetic properties of these objects benefit from both their colloidal nature and the specific properties of the complexes. Hence these new nanocarriers exhibit enhanced T1 MRI contrast, when administered intravenously to mice.


Subject(s)
Contrast Media , Nanostructures , Animals , Mice , Contrast Media/chemistry , Luminescence , Magnetic Resonance Imaging/methods , Polymers , Ions
2.
Nanoscale ; 15(8): 3893-3906, 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36723163

ABSTRACT

Because of the formation of specific antibodies to poly(ethylene glycol) (PEG) leading to life-threatening side effects, there is an increasing need to develop alternatives to treatments and diagnostic methods based on PEGylated copolymers. Block copolymers comprising a poly(N-vinyl-2-pyrrolidone) (PVP) segment can be used for the design of such vectors without any PEG block. As an example, a poly(acrylic acid)-block-poly(N-vinyl-2-pyrrolidone) (PAA-b-PVP) copolymer with controlled composition and molar mass is synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization. Mixing this copolymer with lanthanide cations (Gd3+, Eu3+, Y3+) leads to the formation of hybrid polyion complexes with increased stability, preventing the lanthanide cytotoxicity and in vitro cell penetration. These new nanocarriers exhibit enhanced T1 MRI contrast, when intravenously administered into mice. No leaching of gadolinium ions is detected from such hybrid complexes.


Subject(s)
Contrast Media , Lanthanoid Series Elements , Animals , Mice , Polymers , Magnetic Resonance Imaging , Ions
3.
Front Med (Lausanne) ; 8: 741855, 2021.
Article in English | MEDLINE | ID: mdl-35174180

ABSTRACT

Anti-PDL1 is a monoclonal antibody targeting the programmed death-cell ligand (PD-L1) by blocking the programmed death-cell (PD-1)/PD-L1 axis. It restores the immune system response in several tumours, such as non-small cell lung cancer (NSCLC). Anti-PDL1 or anti-PD1 treatments rely on PD-L1 tumoural expression assessed by immunohistochemistry on biopsy tissue. However, depending on the biopsy extraction site, PD-L1 expression can vary greatly. Non-invasive imaging enables whole-body mapping of PD-L1 sites and could improve the assessment of tumoural PD-L1 expression. METHODS: Pharmacokinetics (PK), biodistribution and dosimetry of a murine anti-PDL1 radiolabelled with zirconium-89, were evaluated in both healthy mice and immunocompetent mice with lung cancer. Preclinical PET (µPET) imaging was used to analyse [89Zr]DFO-Anti-PDL1 distribution in both groups of mice. Non-compartmental (NCA) and compartmental (CA) PK analyses were performed in order to describe PK parameters and assess area under the concentration-time curve (AUC) for dosimetry evaluation in humans. RESULTS: Organ distribution was correctly estimated using PK modelling in both healthy mice and mice with lung cancer. Tumoural uptake occurred within 24 h post-injection of [89Zr]DFO-Anti-PDL1, and the best imaging time was at 48 h according to the signal-to-noise ratio (SNR) and image quality. An in vivo blocking study confirmed that [89Zr]DFO-anti-PDL1 specifically targeted PD-L1 in CMT167 lung tumours in mice. AUC in organs was estimated using a 1-compartment PK model and extrapolated to human (using allometric scaling) in order to estimate the radiation exposure in human. Human-estimated effective dose was 131 µSv/MBq. CONCLUSION: The predicted dosimetry was similar or lower than other antibodies radiolabelled with zirconium-89 for immunoPET imaging.

4.
Cell Rep ; 32(8): 108075, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32846132

ABSTRACT

Atrial natriuretic peptide (ANP) is a cardiac hormone controlling blood volume and pressure in mammals. It is still unclear whether ANP controls cold-induced thermogenesis in vivo. Here, we show that acute cold exposure induces cardiac ANP secretion in mice and humans. Genetic inactivation of ANP promotes cold intolerance and suppresses half of cold-induced brown adipose tissue (BAT) activation in mice. While white adipocytes are resistant to ANP-mediated lipolysis at thermoneutral temperature in mice, cold exposure renders white adipocytes fully responsive to ANP to activate lipolysis and a thermogenic program, a physiological response that is dramatically suppressed in ANP null mice. ANP deficiency also blunts liver triglycerides and glycogen metabolism, thus impairing fuel availability for BAT thermogenesis. ANP directly increases mitochondrial uncoupling and thermogenic gene expression in human white and brown adipocytes. Together, these results indicate that ANP is a major physiological trigger of BAT thermogenesis upon cold exposure in mammals.


Subject(s)
Atrial Natriuretic Factor/metabolism , Thermogenesis/physiology , Animals , Humans , Male , Mice , Mice, Knockout
5.
Methods ; 97: 35-43, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26589188

ABSTRACT

Recently, an increasing number of aptamers have been selected against biomarkers that are expressed at the surface of cells. This class of targets, mostly membrane proteins, is in close contact with the intra- and extra-cellular matrixes and their three-dimensional structures are inextricably linked to their inclusion in lipid bilayers. Therefore, although binding studies can be performed on the isolated form of these proteins, it remains crucial to measure the affinity of these aptamers in a more physiological environment, i.e., directly on living cells. Here, we describe a procedure for radioactive binding assays that can be adapted for measuring the affinity of aptamers against different cell lines. This method has been semi-automated using a liquid handling robot in order to reproducibly measure the apparent dissociation constant Kd and the apparent number of targets per cell. Relevant issues are discussed including the labeling of aptamers, the cells preparation, the incubation, the washings, the use of non-specific competitors, the data analysis and finally the reporting.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques , Membrane Proteins/chemistry , Animals , Cell Adhesion , Humans , PC12 Cells , Protein Binding , Rats , SELEX Aptamer Technique , Sensitivity and Specificity , Single-Cell Analysis
6.
Nucl Med Biol ; 42(8): 643-53, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25963911

ABSTRACT

INTRODUCTION: The N-methyl-D-aspartate receptor (NMDAr) is an ionotropic receptor that mediates excitatory transmission. NMDAr overexcitation is thought to be involved in neurological and neuropsychiatric disorders such as Alzheimer disease and schizophrenia. We synthesized [(18)F]-fluoroethylnormemantine ([(18)F]-FNM), a memantine derivative that binds to phencyclidine (PCP) sites within the NMDA channel pore. These sites are primarily accessible when the channel is in the active and open state. METHODS: Radiosynthesis was carried out using the Raytest® SynChrom R&D fluorination module. Affinity of this new compound was determined by competition assay. We ran a kinetic study in rats and computed a time-activity curve based on a volume-of-interest analysis, using CARIMAS® software. We performed an ex vivo autoradiography, exposing frozen rat brain sections to a phosphorscreen. Adjacent sections were used to detect NMDAr by immunohistochemistry with an anti-NR1 antibody. As a control of the specificity of our compound for NMDAr, we used a rat anesthetized with ketamine. Correlation analysis was performed with ImageJ software between signal of autoradiography and immunostaining. RESULTS: Fluorination yield was 10.5% (end of synthesis), with a mean activity of 3145 MBq and a specific activity above 355 GBq/µmol. Affinity assessment allowed us to determine [(19)F]-FNM IC50 at 6.1 10(-6)M. [(18)F]-FMN concentration gradually increased in the brain, stabilizing at 40 minutes post injection. The brain-to-blood ratio was 6, and 0.4% of the injected dose was found in the brain. Combined ex vivo autoradiography and immunohistochemical staining demonstrated colocalization of NMDAr and [(18)F]-FNM (r=0.622, p<0.0001). The highest intensity was found in the cortex and cerebellum, and the lowest in white matter. A low and homogeneous signal corresponding to unspecific binding was observed when PCP sites were blocked with ketamine. CONCLUSIONS: [(18)F]-FNM appears to be a promising tracer for imaging NMDAr activity for undertaking preclinical studies in perspective of clinical detection of neurological or neuropsychological disorders.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Memantine/analogs & derivatives , Memantine/pharmacokinetics , Phencyclidine/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Autoradiography , Fluorine Radioisotopes/pharmacokinetics , Isotope Labeling , Kinetics , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley , Tissue Distribution
7.
Biochimie ; 94(7): 1595-606, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22738730

ABSTRACT

Nucleic acid Aptamers are ligands that are selected by a process of molecular evolution to bind with high affinities and specificities to a specific target. Recently, an increasing number of aptamers have been selected against biomarkers expressed at the surface of human cells or infectious pathogens. This class of targets, mostly proteins, is associated with several pathologies including cancer, inflammation and infection diseases. Several of these cell surface specific aptamers were tested in vivo as drugs or as targeting agents for nanocarriers, siRNA or contrast agents. Strikingly, they were used to develop a wide variety of new treatments or new approaches for molecular imaging and they were also able to improve current therapies such as chemotherapy, radiotherapy or immunotherapy. This review presents these different applications and the different studies conducted in vivo with this class of aptamers, predominantly in pre-clinical models.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Molecular Imaging/methods , Animals , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Biomarkers/metabolism , Humans
8.
Eur J Nucl Med Mol Imaging ; 37(1): 58-66, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19655139

ABSTRACT

PURPOSE: 6-[(18)F]Fluoro-3,4-dihydroxy-L-phenylalanine (6-[(18)F]F-L-DOPA) is increasingly used for PET imaging of neuroendocrine tumours. In this study, we investigated the use of 6-[(18)F]F-L-DOPA to detect and to monitor the progression of medullary thyroid carcinoma (MTC) in a genetically engineered mouse model of multiple endocrine neoplasia type 2A (MEN2A). METHODS: Dynamic [(18)F]FDG and 6-[(18)F]F-L-DOPA small animal PET scans were acquired during 60 or 90 min in 8- to 20-month-old MEN2A transgenic mice. The kinetics of 6-[(18)F]F-L-DOPA, standardized uptake values (SUV) at 60 min and tumour volumes were recorded. The detection of MTCs using PET was confirmed by autopsy and histological analysis. RESULTS: 6-[(18)F]F-L-DOPA performs better than [(18)F]FDG for MTC detection in this transgenic mouse model. Uptake kinetics of 6-[(18)F]F-L-DOPA in MTCs are very different between mice but, in all cases, high contrast could be observed. Furthermore, 6-[(18)F]F-L-DOPA can detect tumours with sizes (1.8 mm(3)) that are near the resolution limit of PET, whereas they were undetectable by autopsy at the macroscopic level. CONCLUSION: 6-[(18)F]F-L-DOPA PET imaging can monitor the progression of MTCs in a genetically engineered mouse model.


Subject(s)
Carcinoma, Medullary/diagnostic imaging , Dihydroxyphenylalanine/analogs & derivatives , Multiple Endocrine Neoplasia Type 2a/diagnostic imaging , Positron-Emission Tomography/methods , Thyroid Neoplasms/diagnostic imaging , Animals , Mice , Mice, Transgenic , Radiopharmaceuticals , Reproducibility of Results , Sensitivity and Specificity
9.
Blood ; 115(3): 443-52, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-19797522

ABSTRACT

Few techniques are available to characterize in vivo the early cellular dynamics of long-term reconstitution of hematopoiesis after transplantation of hematopoietic stem cells (HSCs) after lethal irradiation. Using a fiber-optic imaging system, we track the early steps of in vivo recruitment and proliferation of Lin(-)Sca-1(+)c-Kit(+)CD34(-) (LSKCD34(-)) HSCs highly enriched in HSCs and transplanted into lethally irradiated mice. Recruitment of the transplanted LSKCD34(-) hematopoietic cells first occurs in the femoral head and is continuous during 24 hours. Quantification of the fluorescence emitted by the transplanted hematopoietic cells shows that proliferation of LSKCD34(-) hematopoietic cells in the femoral head was potent 3 days after transplantation. Using a development of this fiber-optic imaging system, we show that the transplanted LSKCD34(-) hematopoietic cells are associated with vascularized structures as early as 5 hours after transplantation. This early association is dependent on reactive oxygen species (ROS) partly through the regulation of vascular cell adhesion molecule-1 expression on endothelial cells and is followed by a ROS-dependent proliferation of LSKCD34(-) hematopoietic cells. This new in vivo imaging technique permits the observation of the early steps of hematopoietic reconstitution by HSCs in long bones and shows a new role of ROS in the recruitment of HSCs by bone marrow endothelial cells.


Subject(s)
Hematopoiesis/drug effects , Molecular Imaging/methods , Reactive Oxygen Species/pharmacology , Animals , Antigens, CD34/metabolism , Antigens, Ly/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation/drug effects , Femur/cytology , Femur/metabolism , Femur/physiology , Fiber Optic Technology/methods , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Models, Biological , Proto-Oncogene Proteins c-kit/metabolism , Reactive Oxygen Species/metabolism , Validation Studies as Topic , Whole-Body Irradiation
10.
J Cell Mol Med ; 13(9B): 4002-13, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19426156

ABSTRACT

The present study aims at investigating the mechanism by which membrane-type 4 matrix metalloproteinase (MT4-MMP), a membrane-anchored MMP expressed by human breast tumour cells promotes the metastatic dissemination into lung. We applied experimental (intravenous) and spontaneous (subcutaneous) models of lung metastasis using human breast adenocarcinoma MDA-MB-231 cells overexpressing or not MT4-MMP. We found that MT4-MMP does not affect lymph node colonization nor extravasation of cells from the bloodstream, but increases the intravasation step leading to metastasis. Ultrastructural and fluorescent microscopic observations coupled with automatic computer-assisted quantifications revealed that MT4-MMP expression induces blood vessel enlargement and promotes the detachment of mural cells from the vascular tree, thus causing an increased tumour vascular leak. On this basis, we propose that MT4-MMP promotes lung metastasis by disturbing the tumour vessel integrity and thereby facilitating tumour cell intravasation.


Subject(s)
Breast Neoplasms/enzymology , Lung Neoplasms/enzymology , Matrix Metalloproteinase 17/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Line, Tumor , Humans , Lung Neoplasms/secondary , Lymphatic Metastasis , Mammary Neoplasms, Animal/blood supply , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Nude , Microscopy, Electron, Transmission/methods , Microscopy, Fluorescence/methods , Neoplasm Transplantation , Neovascularization, Pathologic , Pericytes/metabolism
11.
Bioconjug Chem ; 19(9): 1921-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18754572

ABSTRACT

We have designed new nanoprobes applicable for both positron emission tomography (PET) and optical fluorescence in vivo imaging. Fluorine-18, which is commonly used for clinical imaging, has been coupled to phospholipid quantum dot (QD) micelles. This probe was injected in mice and we demonstrated that its dynamic quantitative whole body biodistribution and pharmacokinetics could be monitored using PET as well as the kinetics of their cellular uptake using in vivo fibered confocal fluorescence imaging. Phospholipid micelle encapsulation of QDs provides a highly versatile surface chemistry to conjugate multiple chemicals and biomolecules with controlled QD:molecule valency. Here, we show that, in contrast with several previous studies using other QD polymer coatings, these phospholipid QD micelles exhibit long circulation half-time in the bloodstream (on the order of 2 h) and slow uptake by reticulo-endothelial system.


Subject(s)
Diagnostic Imaging/methods , Fluorine Radioisotopes/chemistry , Nanotechnology/methods , Phospholipids/chemistry , Quantum Dots , Whole Body Imaging/methods , Animals , Cadmium Compounds/chemical synthesis , Cell Membrane/metabolism , Kinetics , Mice , Mice, Nude , Micelles , Microscopy, Fluorescence/methods , Nanostructures/chemistry , Positron-Emission Tomography/methods , Selenium Compounds/chemical synthesis
12.
Oligonucleotides ; 16(4): 323-35, 2006.
Article in English | MEDLINE | ID: mdl-17155908

ABSTRACT

Binding of aptamers is dependent on their target conformation, which in turn is conditioned by the target's environment. Therefore, selection of aptamers against the active forms of membrane proteins could require their correct membrane insertion in order to maintain their native conformation. Here, we compare different SELEX strategies to identify aptamers against the mutated form of the membrane receptor tyrosine kinase RET(C634Y). (1) selections S1 and S2 against living cells transformed to express the protein yielded a minority of RET-targeted aptamers while the bulk of aptamers recognized more abundant membrane proteins, suggesting that a high level of expression of the target protein is crucial to allow the isolation of aptamers at cell surface; (2) selection S3 against the purified extracellular moiety of RET yielded aptamers unable to recognize RET expressed at the cell membrane; (3) crossover selections S4 and S5 alternating cells and recombinant RET enhanced the enrichment of the aptamers directed against RET; however, these aptamers displayed a weaker affinity for Ret than those obtained with S1 and S2. In our case, using transformed cell lines as the partitioning matrix during SELEX appears to be essential in order to obtain aptamers able to recognize the RET receptor tyrosine kinase in its physiologic environment.


Subject(s)
Aptamers, Nucleotide/isolation & purification , Aptamers, Nucleotide/metabolism , Membrane Proteins/metabolism , SELEX Aptamer Technique/methods , Amino Acid Substitution , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Base Sequence , Cross-Linking Reagents , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleic Acid Conformation , PC12 Cells , Polymorphism, Restriction Fragment Length , Protein Binding , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
Mol Cancer Res ; 4(7): 481-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16849523

ABSTRACT

In several neuroblastoma cell lines, retinoic acid (RA)-induced differentiation is coupled to increased expression of functional neurotrophic factor receptors, including Trk family receptors and the glial cell-derived neurotrophic factor receptor, Ret. In several cases, increased expression is dependent on signaling through TrkB. Unlike TrkA and TrkB, Ret has never been implicated as a prognostic marker for neuroblastomas. SK-N-BE(2) cells do not express any of Trk family receptors; therefore, they are a choice system to study the specific role of Ret in RA-induced differentiation. Using a 2'-fluoro-RNA aptamer and a truncated Ret protein as specific inhibitors of Ret, we show that RA-induced differentiation is mediated by a positive autocrine loop that sustains Ret downstream signaling and depends on glial cell-derived neurotrophic factor expression and release. This report shows that in SK-N-BE(2) cells, stimulation of Ret is a major upstream mechanism needed to mediate RA-induced differentiation. These results provide important insights on the molecular mechanism of RA action, which might be relevant for the development of biologically based therapeutic strategies.


Subject(s)
Cell Differentiation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Tretinoin/pharmacology , Animals , Autocrine Communication/drug effects , Cell Line, Tumor , Dimerization , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Neuroblastoma/pathology
14.
PLoS Biol ; 3(4): e123, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15769183

ABSTRACT

Targeting large transmembrane molecules, including receptor tyrosine kinases, is a major pharmacological challenge. Specific oligonucleotide ligands (aptamers) can be generated for a variety of targets through the iterative evolution of a random pool of sequences (SELEX). Nuclease-resistant aptamers that recognize the human receptor tyrosine kinase RET were obtained using RET-expressing cells as targets in a modified SELEX procedure. Remarkably, one of these aptamers blocked RET-dependent intracellular signaling pathways by interfering with receptor dimerization when the latter was induced by the physiological ligand or by an activating mutation. This strategy is generally applicable to transmembrane receptors and opens the way to targeting other members of this class of proteins that are of major biomedical importance.


Subject(s)
Proto-Oncogene Proteins c-ret/genetics , SELEX Aptamer Technique/methods , 3T3 Cells , Amino Acid Substitution , Animals , Base Sequence , Chlorocebus aethiops , Enzyme Inhibitors , Humans , Mice , Molecular Sequence Data , Multiple Endocrine Neoplasia Type 2b/genetics , PC12 Cells , Pheochromocytoma , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Rats , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...