Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Health Phys ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905438

ABSTRACT

ABSTRACT: During a nuclear/radiological incident or an accident involving internal intakes with radioactive cobalt or strontium, the recommended treatments, consisting of the administration of diethylenetriaminepentaacetic acid for 60 Co and calcium gluconate for 90 Sr, are of low specificity, and their effectiveness can be enhanced. In this manuscript, a liposomal formulation was developed to deliver potential chelating agents to the main retention organs of both radionuclides. A bisphosphonate, etidronate, has been selected as a possible candidate due to its satisfying decorporation activity for uranium, bone tropism, and potential affinity with cobalt. Pre-clinical studies have been carried out on rats using radionuclide contamination and treatment administration by the intravenous route. The effectiveness of free or liposomal etidronate was evaluated, with an administration at 30 min, 48 h post-contamination with 60 Co. Regarding 85 Sr, a more extended experiment with etidronate liposomes was performed over 6 d. The results were compared to those performed with reference treatments, diethylenetriaminepentaacetic acid for cobalt and calcium gluconate for strontium. Unexpected results were found for the reference treatments that were significantly less effective than previously reported or showed no effectiveness. Free etidronate revealed no significant efficacy after 48 h, but the liposomal form suggested an interaction with radionuclides, not sufficient to change the biokinetics. This study emphasizes the need for early treatment administration and further research to provide a more effective medical countermeasure.

2.
Arthritis Rheumatol ; 76(1): 18-31, 2024 01.
Article in English | MEDLINE | ID: mdl-37527031

ABSTRACT

OBJECTIVE: We previously reported an increased expression of microRNA-155 (miR-155) in the blood monocytes of patients with rheumatoid arthritis (RA) that could be responsible for impaired monocyte polarization to anti-inflammatory M2-like macrophages. In this study, we employed two preclinical models of RA, collagen-induced arthritis and K/BxN serum transfer arthritis, to examine the therapeutic potential of antagomiR-155-5p entrapped within PEGylated (polyethylene glycol [PEG]) liposomes in resolution of arthritis and repolarization of monocytes towards the anti-inflammatory M2 phenotype. METHODS: AntagomiR-155-5p or antagomiR-control were encapsulated in PEG liposomes of 100 nm in size and -10 mV in zeta potential with high antagomiR loading efficiency (above 80%). Mice were injected intravenously with 1.5 nmol/100 µL PEG liposomes containing antagomiR-155-5p or control after the induction of arthritis. RESULTS: We demonstrated the biodistribution of fluorescently tagged PEG liposomes to inflamed joints one hour after the injection of fluorescently tagged PEG liposomes, as well as the liver's subsequent accumulation after 48 hours, indicative of hepatic clearance, in mice with arthritis. The injection of PEG liposomes containing antagomiR-155-5p decreased arthritis score and paw swelling compared with PEG liposomes containing antagomiR-control or the systemic delivery of free antagomiR-155-5p. Moreover, treatment with PEG liposomes containing antagomiR-155-5p led to the restoration of bone marrow monocyte defects in anti-inflammatory macrophage differentiation without any significant functional change in other immune cells, including splenic B and T cells. CONCLUSION: The injection of antagomiR-155-5p encapsulated in PEG liposomes allows the delivery of small RNA to monocytes and macrophages and reduces joint inflammation in murine models of RA, providing a promising strategy in human disease.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , MicroRNAs , Humans , Mice , Animals , Antagomirs/metabolism , Antagomirs/therapeutic use , Liposomes/metabolism , Liposomes/therapeutic use , Tissue Distribution , Macrophages , Anti-Inflammatory Agents/therapeutic use , MicroRNAs/metabolism
3.
Int J Pharm ; 639: 122946, 2023 May 25.
Article in English | MEDLINE | ID: mdl-37044230

ABSTRACT

Dexamethasone is a well-known anti-inflammatory drug readily used to treat many lung diseases. However, its side effects and poor lower airway deposition and retention are significant limitations to its usage. In this work, we developed lipid nanoparticulate platforms loaded with dexamethasone and evaluated their behavior in inflammatory lung models in vitro and in vivo. Dexamethasone-loaded liposomes with an average diameter below 150 nm were obtained using a solvent injection method. Three different formulations were produced with a distinct surface coating (polyethylene glycol, hyaluronic acid, or a mixture of both) as innovative strategies to cross the pulmonary mucus layer and/or target CD44 expressed on alveolar proinflammatory macrophages. Interestingly, while electron paramagnetic spectroscopy showed that surface modifications did not induce any molecular changes in the liposomal membrane, drug loading analysis revealed that adding the hyaluronic acid in the bilayer led to a decrease of dexamethasone loading (from 3.0 to 1.7 w/w%). In vitro experiments on LPS-activated macrophages demonstrated that the encapsulation of dexamethasone in liposomes, particularly in HA-bearing ones, improved its anti-inflammatory efficacy compared to the free drug. Subsequently, in vivo data revealed that while intratracheal administration of free dexamethasone led to an important inter-animals variation of efficacy, dexamethasone-loaded liposomes showed an improved consistency within the results. Our data indicate that encapsulating dexamethasone into lipid nanoparticles is a potent strategy to improve its efficacy after lung delivery.


Subject(s)
Hyaluronic Acid , Liposomes , Animals , Liposomes/chemistry , Hyaluronic Acid/chemistry , Anti-Inflammatory Agents , Macrophages , Dexamethasone
4.
Biomacromolecules ; 24(2): 667-677, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36599673

ABSTRACT

Small interfering RNA (siRNA) holds promise for treating rheumatoid arthritis by inhibiting major cytokines such as tumor necrosis factor-α (TNF-α). We developed original cationic amphiphilic phosphorus dendrons to produce dendriplexes associated with TNF-α siRNA. The dendrons were made of 10 pyrrolidinium end groups and a C17 aliphatic chain. The dendriplexes demonstrated the ability to protect siRNA from nuclease degradation and to promote macrophage uptake. Moreover, they led to potent inhibition of TNF-α expression in the lipopolysaccharide-activated mouse macrophage cell line RAW264.7 in vitro model. A significant anti-inflammatory effect in the murine collagen-induced arthritis model was observed through arthritis scoring and histological observations. These results open up essential perspectives in using this original amphiphilic dendron to reduce the disease burden and improve outcomes in chronic inflammatory diseases.


Subject(s)
Arthritis, Experimental , Dendrimers , Animals , Mice , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tumor Necrosis Factor-alpha/genetics , Anti-Inflammatory Agents/pharmacology
5.
J Control Release ; 352: 15-24, 2022 12.
Article in English | MEDLINE | ID: mdl-36209941

ABSTRACT

Alveolar macrophages play a crucial role in the initiation and resolution of the immune response in the lungs. Pro-inflammatory M1 alveolar macrophages are an interesting target for treating inflammatory and infectious pulmonary diseases. One commune targeting strategy is to use nanoparticles conjugated with hyaluronic acid, which interact with CD44 overexpressed on the membrane of those cells. Unfortunately, this coating strategy may be countered by the presence on the surface of the nanoparticles of a poly(ethylene glycol) corona employed to improve nanoparticles' diffusion in the lung mucus. This study aims to measure this phenomenon by comparing the behavior in a murine lung inflammation model of three liposomal platforms designed to represent different poly(ethylene glycol) and hyaluronic acid densities (Liposome-PEG, Liposome-PEG-HA and Liposome-HA). In this work, the liposomes were obtained by a one-step ethanol injection method. Their interaction with mucin and targeting ability toward pro-inflammatory macrophages were then investigated in vitro and in vivo in a LPS model of lung inflammation. In vitro, poly(ethylene glycol) free HA-liposomes display a superior targeting efficiency toward M1 macrophages, while the addition of poly(ethylene glycol) induces better mucus mobility. Interestingly in vivo studies revealed that the three liposomes showed distinct cell specificity with alveolar macrophages demonstrating an avidity for poly(ethylene glycol) free HA-liposomes, while neutrophils favored PEGylated liposomes exempt of HA. Those results could be explained by the presence of two forces exercising a balance between mucus penetration and receptor targeting. This study corroborates the importance of considering the site of action and the targeted cells when designing nanoparticles to treat lung diseases.


Subject(s)
Hyaluronic Acid , Liposomes , Mice , Animals , Macrophages, Alveolar , Polyethylene Glycols , Mucus
6.
Adv Drug Deliv Rev ; 175: 113809, 2021 08.
Article in English | MEDLINE | ID: mdl-34033819

ABSTRACT

Thanks to their abilities to modulate the expression of virtually any genes, RNA therapeutics have attracted considerable research efforts. Among the strategies focusing on nucleic acid gene inhibitors, antisense oligonucleotides and small interfering RNAs have reached advanced clinical trial phases with several of them having recently been marketed. These successes were obtained by overcoming stability and cellular delivery issues using either chemically modified nucleic acids or nanoparticles. As nucleic acid gene inhibitors are promising strategies to treat inflammatory diseases, this review focuses on the barriers, from manufacturing issues to cellular/subcellular delivery, that still need to be overcome to deliver the nucleic acids to sites of inflammation other than the liver. Furthermore, key examples of applications in rheumatoid arthritis, inflammatory bowel, and lung diseases are presented as case studies of systemic, oral, and lung nucleic acid delivery.


Subject(s)
Inflammation/drug therapy , Nanomedicine/methods , Nanoparticle Drug Delivery System , Nucleic Acids/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Drug Delivery Systems/methods , Genes/drug effects , Humans , Inflammation/genetics , Nucleic Acids/therapeutic use , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use
7.
Sci Transl Med ; 13(584)2021 03 10.
Article in English | MEDLINE | ID: mdl-33692130

ABSTRACT

Macrophages play a central role in the pathogenesis of atherosclerosis. The inflammatory properties of these cells are dictated by their metabolism, of which the mechanistic target of rapamycin (mTOR) signaling pathway is a key regulator. Using myeloid cell-specific nanobiologics in apolipoprotein E-deficient (Apoe -/-) mice, we found that targeting the mTOR and ribosomal protein S6 kinase-1 (S6K1) signaling pathways rapidly diminished plaque macrophages' inflammatory activity. By investigating transcriptome modifications, we identified Psap, a gene encoding the lysosomal protein prosaposin, as closely related with mTOR signaling. Subsequent in vitro experiments revealed that Psap inhibition suppressed both glycolysis and oxidative phosphorylation. Transplantation of Psap -/- bone marrow to low-density lipoprotein receptor knockout (Ldlr -/-) mice led to a reduction in atherosclerosis development and plaque inflammation. Last, we confirmed the relationship between PSAP expression and inflammation in human carotid atherosclerotic plaques. Our findings provide mechanistic insights into the development of atherosclerosis and identify prosaposin as a potential therapeutic target.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Saposins/therapeutic use , Animals , Disease Models, Animal , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE
8.
Nat Nanotechnol ; 15(5): 398-405, 2020 05.
Article in English | MEDLINE | ID: mdl-32313216

ABSTRACT

Ischaemic heart disease evokes a complex immune response. However, tools to track the systemic behaviour and dynamics of leukocytes non-invasively in vivo are lacking. Here, we present a multimodal hot-spot imaging approach using an innovative high-density lipoprotein-derived nanotracer with a perfluoro-crown ether payload (19F-HDL) to allow myeloid cell tracking by 19F magnetic resonance imaging. The 19F-HDL nanotracer can additionally be labelled with zirconium-89 and fluorophores to detect myeloid cells by in vivo positron emission tomography imaging and optical modalities, respectively. Using our nanotracer in atherosclerotic mice with myocardial infarction, we observed rapid myeloid cell egress from the spleen and bone marrow by in vivo 19F-HDL magnetic resonance imaging. Concurrently, using ex vivo techniques, we showed that circulating pro-inflammatory myeloid cells accumulated in atherosclerotic plaques and at the myocardial infarct site. Our multimodality imaging approach is a valuable addition to the immunology toolbox, enabling the study of complex myeloid cell behaviour dynamically.


Subject(s)
Myeloid Cells/pathology , Myocardial Ischemia/diagnostic imaging , Animals , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , Cell Tracking/methods , Crown Ethers/analysis , Female , Fluorescent Dyes/analysis , Fluorine/analysis , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred C57BL , Multimodal Imaging/methods , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Optical Imaging/methods , Positron-Emission Tomography/methods , Radioisotopes/analysis , Zirconium/analysis
9.
Bioconjug Chem ; 31(2): 360-368, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31095372

ABSTRACT

Nanotherapy has recently emerged as an experimental treatment option for atherosclerosis. To fulfill its promise, robust noninvasive imaging approaches for subject selection and treatment evaluation are warranted. To that end, we present here a positron emission tomography (PET)-based method for quantification of liposomal nanoparticle uptake in the atherosclerotic vessel wall. We evaluated a modular procedure to label liposomal nanoparticles with the radioisotope zirconium-89 (89Zr). Their biodistribution and vessel wall targeting in a rabbit atherosclerosis model was evaluated up to 15 days after intravenous injection by PET/computed tomography (CT) and PET/magnetic resonance imaging (PET/MRI). Vascular permeability was assessed in vivo using three-dimensional dynamic contrast-enhanced MRI (3D DCE-MRI) and ex vivo using near-infrared fluorescence (NIRF) imaging. The 89Zr-radiolabeled liposomes displayed a biodistribution pattern typical of long-circulating nanoparticles. Importantly, they markedly accumulated in atherosclerotic lesions in the abdominal aorta, as evident on PET/MRI and confirmed by autoradiography, and this uptake moderately correlated with vascular permeability. The method presented herein facilitates the development of nanotherapy for atherosclerotic disease as it provides a tool to screen for nanoparticle targeting in individual subjects' plaques.


Subject(s)
Atherosclerosis/diagnostic imaging , Liposomes/analysis , Plaque, Atherosclerotic/diagnostic imaging , Positron-Emission Tomography/methods , Radioisotopes/analysis , Zirconium/analysis , Animals , Aorta, Abdominal/diagnostic imaging , Male , Rabbits , Tissue Distribution
10.
Sci Transl Med ; 11(506)2019 08 21.
Article in English | MEDLINE | ID: mdl-31434756

ABSTRACT

Nanomedicine research produces hundreds of studies every year, yet very few formulations have been approved for clinical use. This is due in part to a reliance on murine studies, which have limited value in accurately predicting translational efficacy in larger animal models and humans. Here, we report the scale-up of a nanoimmunotherapy from mouse to large rabbit and porcine atherosclerosis models, with an emphasis on the solutions we implemented to overcome production and evaluation challenges. Specifically, we integrated translational imaging readouts within our workflow to both analyze the nanoimmunotherapeutic's in vivo behavior and assess treatment response in larger animals. We observed our nanoimmunotherapeutic's anti-inflammatory efficacy in mice, as well as rabbits and pigs. Nanoimmunotherapy-mediated reduction of inflammation in the large animal models halted plaque progression, supporting the approach's translatability and potential to acutely treat atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/therapy , Imaging, Three-Dimensional , Immunotherapy , Nanomedicine , Animals , Apolipoproteins E/deficiency , Atherosclerosis/diagnostic imaging , Atherosclerosis/drug therapy , Disease Models, Animal , Female , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/toxicity , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Positron-Emission Tomography , Rabbits , Simvastatin/pharmacology , Simvastatin/therapeutic use , Species Specificity , Swine , Tissue Distribution
11.
JACC Cardiovasc Imaging ; 12(10): 2015-2026, 2019 10.
Article in English | MEDLINE | ID: mdl-30343086

ABSTRACT

OBJECTIVES: This study sought to develop an integrative positron emission tomography (PET) with magnetic resonance imaging (MRI) procedure for accurate atherosclerotic plaque phenotyping, facilitated by clinically approved and nanobody radiotracers. BACKGROUND: Noninvasive characterization of atherosclerosis remains a challenge in clinical practice. The limitations of current diagnostic methods demonstrate that, in addition to atherosclerotic plaque morphology and composition, disease activity needs to be evaluated. METHODS: We screened 3 nanobody radiotracers targeted to different biomarkers of atherosclerosis progression, namely vascular cell adhesion molecule (VCAM)-1, lectin-like oxidized low-density lipoprotein receptor (LOX)-1, and macrophage mannose receptor (MMR). The nanobodies, initially radiolabeled with copper-64 (64Cu), were extensively evaluated in Apoe-/- mice and atherosclerotic rabbits using a combination of in vivo PET/MRI readouts and ex vivo radioactivity counting, autoradiography, and histological analyses. RESULTS: The 3 nanobody radiotracers accumulated in atherosclerotic plaques and displayed short circulation times due to fast renal clearance. The MMR nanobody was selected for labeling with gallium-68 (68Ga), a short-lived radioisotope with high clinical relevance, and used in an ensuing atherosclerosis progression PET/MRI study. Macrophage burden was longitudinally studied by 68Ga-MMR-PET, plaque burden by T2-weighted MRI, and neovascularization by dynamic contrast-enhanced (DCE) MRI. Additionally, inflammation and microcalcifications were evaluated by fluorine-18 (18F)-labeled fluorodeoxyglucose (18F-FDG) and 18F-sodium fluoride (18F-NaF) PET, respectively. We observed an increase in all the aforementioned measures as disease progressed, and the imaging signatures correlated with histopathological features. CONCLUSIONS: We have evaluated nanobody-based radiotracers in rabbits and developed an integrative PET/MRI protocol that allows noninvasive assessment of different processes relevant to atherosclerosis progression. This approach allows the multiparametric study of atherosclerosis and can aid in early stage anti-atherosclerosis drug trials.


Subject(s)
Atherosclerosis/diagnostic imaging , Multiparametric Magnetic Resonance Imaging , Plaque, Atherosclerotic , Positron-Emission Tomography , Radiopharmaceuticals/administration & dosage , Single-Domain Antibodies/administration & dosage , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Disease Models, Animal , Disease Progression , Early Diagnosis , Genetic Predisposition to Disease , Lectins, C-Type/immunology , Mannose Receptor , Mannose-Binding Lectins/immunology , Mice, Knockout, ApoE , Multimodal Imaging , Phenotype , Rabbits , Radiopharmaceuticals/pharmacokinetics , Receptors, Cell Surface/immunology , Scavenger Receptors, Class E/immunology , Single-Domain Antibodies/metabolism , Vascular Cell Adhesion Molecule-1/immunology
12.
Immunity ; 49(5): 819-828.e6, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413362

ABSTRACT

Inducing graft acceptance without chronic immunosuppression remains an elusive goal in organ transplantation. Using an experimental transplantation mouse model, we demonstrate that local macrophage activation through dectin-1 and toll-like receptor 4 (TLR4) drives trained immunity-associated cytokine production during allograft rejection. We conducted nanoimmunotherapeutic studies and found that a short-term mTOR-specific high-density lipoprotein (HDL) nanobiologic treatment (mTORi-HDL) averted macrophage aerobic glycolysis and the epigenetic modifications underlying inflammatory cytokine production. The resulting regulatory macrophages prevented alloreactive CD8+ T cell-mediated immunity and promoted tolerogenic CD4+ regulatory T (Treg) cell expansion. To enhance therapeutic efficacy, we complemented the mTORi-HDL treatment with a CD40-TRAF6-specific nanobiologic (TRAF6i-HDL) that inhibits co-stimulation. This synergistic nanoimmunotherapy resulted in indefinite allograft survival. Together, we show that HDL-based nanoimmunotherapy can be employed to control macrophage function in vivo. Our strategy, focused on preventing inflammatory innate immune responses, provides a framework for developing targeted therapies that promote immunological tolerance.


Subject(s)
Graft Survival/immunology , Immunosuppression Therapy , Inflammation/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Organ Transplantation , Allografts , Animals , Biomarkers , HMGB1 Protein/genetics , Immune Tolerance , Immunity, Innate , Immunologic Memory , Macrophages/immunology , Macrophages/metabolism , Mice , TOR Serine-Threonine Kinases/metabolism , Vimentin/genetics
13.
Methods Mol Biol ; 1816: 385-400, 2018.
Article in English | MEDLINE | ID: mdl-29987836

ABSTRACT

Several animal models have been developed to study atherosclerosis. Here we present a rabbit atherosclerosis model generated by surgical denudation of the aortic endothelium in combination with a high-fat and cholesterol-enriched diet. This model is characterized by the formation of vascular lesions that exhibit several hallmarks of human atherosclerosis. Due to the rabbit's relative large size, as compared to rodents, this model is suited for the imaging-guided evaluation of novel therapeutic strategies using clinical scanners. In this chapter, we present an extensive outline of the procedures to induce aortic atherosclerotic lesions in rabbits as well as methods to evaluate the disease, including noninvasive in vivo multiparametric imaging and histopathology.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/pathology , Disease Models, Animal , Rabbits , Animals , Aorta/diagnostic imaging , Aorta/pathology , Atherosclerosis/diagnostic imaging , Diet, High-Fat/adverse effects , Humans , Magnetic Resonance Imaging/methods , Male , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/pathology , Positron-Emission Tomography/methods , Rabbits/physiology
14.
J Am Coll Cardiol ; 71(5): 527-542, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29406859

ABSTRACT

BACKGROUND: Disrupting the costimulatory CD40-CD40L dyad reduces atherosclerosis, but can result in immune suppression. The authors recently identified small molecule inhibitors that block the interaction between CD40 and tumor necrosis factor receptor-associated factor (TRAF) 6 (TRAF-STOPs), while leaving CD40-TRAF2/3/5 interactions intact, thereby preserving CD40-mediated immunity. OBJECTIVES: This study evaluates the potential of TRAF-STOP treatment in atherosclerosis. METHODS: The effects of TRAF-STOPs on atherosclerosis were investigated in apolipoprotein E deficient (Apoe-/-) mice. Recombinant high-density lipoprotein (rHDL) nanoparticles were used to target TRAF-STOPs to macrophages. RESULTS: TRAF-STOP treatment of young Apoe-/- mice reduced atherosclerosis by reducing CD40 and integrin expression in classical monocytes, thereby hampering monocyte recruitment. When Apoe-/- mice with established atherosclerosis were treated with TRAF-STOPs, plaque progression was halted, and plaques contained an increase in collagen, developed small necrotic cores, and contained only a few immune cells. TRAF-STOP treatment did not impair "classical" immune pathways of CD40, including T-cell proliferation and costimulation, Ig isotype switching, or germinal center formation, but reduced CD40 and ß2-integrin expression in inflammatory monocytes. In vitro testing and transcriptional profiling showed that TRAF-STOPs are effective in reducing macrophage migration and activation, which could be attributed to reduced phosphorylation of signaling intermediates of the canonical NF-κB pathway. To target TRAF-STOPs specifically to macrophages, TRAF-STOP 6877002 was incorporated into rHDL nanoparticles. Six weeks of rHDL-6877002 treatment attenuated the initiation of atherosclerosis in Apoe-/- mice. CONCLUSIONS: TRAF-STOPs can overcome the current limitations of long-term CD40 inhibition in atherosclerosis and have the potential to become a future therapeutic for atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Atherosclerosis/prevention & control , CD40 Ligand/antagonists & inhibitors , Macrophages/drug effects , Signal Transduction/drug effects , TNF Receptor-Associated Factor 6/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Cell Culture Techniques , Cell Movement/drug effects , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Propiophenones/pharmacology
15.
Am J Transplant ; 18(5): 1247-1255, 2018 05.
Article in English | MEDLINE | ID: mdl-29314558

ABSTRACT

The colony-stimulating factor 1 (CSF1) regulates the differentiation and function of tissue macrophages and determines the outcome of the immune response. The molecular mechanisms behind CSF1-mediated macrophage development remain to be elucidated. Here we demonstrate that neutrophil-derived CSF1 controls macrophage polarization and proliferation, which is necessary for the induction of tolerance. Inhibiting neutrophil production of CSF1 or preventing macrophage proliferation, using targeted nanoparticles loaded with the cell cycle inhibitor simvastatin, abrogates the induction of tolerance. These results provide new mechanistic insights into the developmental requirements of tolerogenic macrophages and identify CSF1 producing neutrophils as critical regulators of the immunological response.


Subject(s)
Heart Transplantation , Immune Tolerance/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Neutrophils/immunology , Transplantation Tolerance/immunology , Animals , Cell Differentiation , Cell Proliferation , Female , Humans , Macrophages/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/metabolism , Signal Transduction
16.
J Am Coll Cardiol ; 71(3): 321-335, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29348025

ABSTRACT

BACKGROUND: Oxidation-specific epitopes (OSEs) are proinflammatory, and elevated levels in plasma predict cardiovascular events. OBJECTIVES: The purpose of this study was to develop novel positron emission tomography (PET) probes to noninvasively image OSE-rich lesions. METHODS: An antigen-binding fragment (Fab) antibody library was constructed from human fetal cord blood. After multiple rounds of screening against malondialdehyde-acetaldehyde (MAA) epitopes, the Fab LA25 containing minimal nontemplated insertions in the CDR3 region was identified and characterized. In mice, pharmacokinetics, biodistribution, and plaque specificity studies were performed with Zirconium-89 (89Zr)-labeled LA25. In rabbits, 89Zr-LA25 was used in combination with an integrated clinical PET/magnetic resonance (MR) system. 18F-fluorodeoxyglucose PET and dynamic contrast-enhanced MR imaging were used to evaluate vessel wall inflammation and plaque neovascularization, respectively. Extensive ex vivo validation was carried out through a combination of gamma counting, near infrared fluorescence, autoradiography, immunohistochemistry, and immunofluorescence. RESULTS: LA25 bound specifically to MAA epitopes in advanced and ruptured human atherosclerotic plaques with accompanying thrombi and in debris from distal protection devices. PET/MR imaging 24 h after injection of 89Zr-LA25 showed increased uptake in the abdominal aorta of atherosclerotic rabbits compared with nonatherosclerotic control rabbits, confirmed by ex vivo gamma counting and autoradiography. 18F-fluorodeoxyglucose PET, dynamic contrast-enhanced MR imaging, and near-infrared fluorescence signals were also significantly higher in atherosclerotic rabbit aortas compared with control aortas. Enhanced liver uptake was also noted in atherosclerotic animals, confirmed by the presence of MAA epitopes by immunostaining. CONCLUSIONS: 89Zr-LA25 is a novel PET radiotracer that may allow noninvasive phenotyping of high-risk OSE-rich lesions.


Subject(s)
Acetaldehyde , Atherosclerosis/diagnostic imaging , Magnetic Resonance Imaging/methods , Malondialdehyde , Positron-Emission Tomography/methods , Thrombosis/diagnostic imaging , Acetaldehyde/metabolism , Animals , Atherosclerosis/metabolism , Epitopes/metabolism , Humans , Malondialdehyde/metabolism , Mice , Mice, Knockout , Rabbits , Thrombosis/metabolism , Tissue Distribution/physiology
17.
Nat Biomed Eng ; 2(5): 279-292, 2018 05.
Article in English | MEDLINE | ID: mdl-30936448

ABSTRACT

Macrophage accumulation in atherosclerosis is directly linked to the destabilization and rupture of plaque, causing acute atherothrombotic events. Circulating monocytes enter the plaque and differentiate into macrophages, where they are activated by CD4+ T lymphocytes through CD40-CD40 ligand signalling. Here, we report the development and multiparametric evaluation of a nanoimmunotherapy that moderates CD40-CD40 ligand signalling in monocytes and macrophages by blocking the interaction between CD40 and tumour necrosis factor receptor-associated factor 6 (TRAF6). We evaluated the biodistribution characteristics of the nanoimmunotherapy in apolipoprotein E-deficient (Apoe-/-) mice and in non-human primates by in vivo positron-emission tomography imaging. In Apoe-/- mice, a 1-week nanoimmunotherapy treatment regimen achieved significant anti-inflammatory effects, which was due to the impaired migration capacity of monocytes, as established by a transcriptome analysis. The rapid reduction of plaque inflammation by the TRAF6-targeted nanoimmunotherapy and its favourable toxicity profiles in both mice and non-human primates highlights the translational potential of this strategy for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/therapy , Immunotherapy/methods , Nanomedicine/methods , TNF Receptor-Associated Factor 6/metabolism , Animals , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Disease Models, Animal , Female , Macaca fascicularis , Macrophages/immunology , Male , Mice , Mice, Transgenic , Monocytes/immunology , TNF Receptor-Associated Factor 6/chemistry , Tissue Distribution
19.
J Control Release ; 262: 47-57, 2017 Sep 28.
Article in English | MEDLINE | ID: mdl-28700897

ABSTRACT

Atherosclerosis is a leading cause of worldwide morbidity and mortality whose management could benefit from novel targeted therapeutics. Nanoparticles are emerging as targeted drug delivery systems in chronic inflammatory disorders. To optimally exploit nanomedicines, understanding their biological behavior is crucial for further development of clinically relevant and efficacious nanotherapeutics intended to reduce plaque inflammation. Here, three clinically relevant nanomedicines, i.e., high-density lipoprotein ([S]-HDL), polymeric micelles ([S]-PM), and liposomes ([S]-LIP), that are loaded with the HMG-CoA reductase inhibitor simvastatin [S], were evaluated in the apolipoprotein E-deficient (Apoe-/-) mouse model of atherosclerosis. We systematically employed quantitative techniques, including in vivo positron emission tomography imaging, gamma counting, and flow cytometry to evaluate the biodistribution, nanomedicines' uptake by plaque-associated macrophages/monocytes, and their efficacy to reduce macrophage burden in atherosclerotic plaques. The three formulations demonstrated distinct biological behavior in Apoe-/- mice. While [S]-PM and [S]-LIP possessed longer circulation half-lives, the three platforms accumulated to similar levels in atherosclerotic plaques. Moreover, [S]-HDL and [S]-PM showed higher uptake by plaque macrophages in comparison to [S]-LIP, while [S]-PM demonstrated the highest uptake by Ly6Chigh monocytes. Among the three formulations, [S]-PM displayed the highest efficacy in reducing macrophage burden in advanced atherosclerotic plaques. In conclusion, our data demonstrate that [S]-PM is a promising targeted drug delivery system, which can be advanced for the treatment of atherosclerosis and other inflammatory disorders in the clinical settings. Our results also emphasize the importance of a thorough understanding of nanomedicines' biological performance, ranging from the whole body to the target cells, as well drug retention in the nanoparticles. Such systematic investigations would allow rational applications of nanomaterials', beyond cancer, facilitating the expansion of the nanomedicine horizon.


Subject(s)
Atherosclerosis/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Simvastatin/administration & dosage , Animals , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Carbocyanines/administration & dosage , Carbocyanines/pharmacokinetics , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipoproteins, HDL/administration & dosage , Lipoproteins, HDL/pharmacokinetics , Liposomes , Mice, Knockout , Micelles , Nanomedicine , Radioisotopes , Simvastatin/blood , Simvastatin/pharmacokinetics , Simvastatin/therapeutic use , Zirconium
20.
ACS Nano ; 11(6): 5785-5799, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28463501

ABSTRACT

Hyaluronan is a biologically active polymer, which can be formulated into nanoparticles. In our study, we aimed to probe atherosclerosis-associated inflammation by using hyaluronan nanoparticles and to determine whether they can ameliorate atherosclerosis. Hyaluronan nanoparticles (HA-NPs) were prepared by reacting amine-functionalized oligomeric hyaluronan (HA) with cholanic ester and labeled with a fluorescent or radioactive label. HA-NPs were characterized in vitro by several advanced microscopy methods. The targeting properties and biodistribution of HA-NPs were studied in apoe-/- mice, which received either fluorescent or radiolabeled HA-NPs and were examined ex vivo by flow cytometry or nuclear techniques. Furthermore, three atherosclerotic rabbits received 89Zr-HA-NPs and were imaged by PET/MRI. The therapeutic effects of HA-NPs were studied in apoe-/- mice, which received weekly doses of 50 mg/kg HA-NPs during a 12-week high-fat diet feeding period. Hydrated HA-NPs were ca. 90 nm in diameter and displayed very stable morphology under hydrolysis conditions. Flow cytometry revealed a 6- to 40-fold higher uptake of Cy7-HA-NPs by aortic macrophages compared to normal tissue macrophages. Interestingly, both local and systemic HA-NP-immune cell interactions significantly decreased over the disease progression. 89Zr-HA-NPs-induced radioactivity in atherosclerotic aortas was 30% higher than in wild-type controls. PET imaging of rabbits revealed 6-fold higher standardized uptake values compared to the muscle. The plaques of HA-NP-treated mice contained 30% fewer macrophages compared to control and free HA-treated group. In conclusion, we show favorable targeting properties of HA-NPs, which can be exploited for PET imaging of atherosclerosis-associated inflammation. Furthermore, we demonstrate the anti-inflammatory effects of HA-NPs in atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , Hyaluronic Acid/therapeutic use , Macrophages/drug effects , Nanoparticles/therapeutic use , Plaque, Atherosclerotic/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacokinetics , Macrophages/pathology , Male , Mice , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/pathology , Positron-Emission Tomography , Rabbits , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...