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










Publication year range
1.
Clin Transl Sci ; 15(1): 244-254, 2022 01.
Article in English | MEDLINE | ID: mdl-34416076

ABSTRACT

This work presents an end-to-end approach for assessing the absolute bioavailability of highly hydrophobic, poorly water-soluble compounds that exhibit high nonspecific binding using venetoclax as a model drug. The approach utilizes a stable labeled i.v. microdose and requires fewer resources compared with traditional approaches that use radioactive 14 C-labeled compounds. The stable labeled venetoclax and internal standard were synthesized, then an i.v. formulation was developed. In the clinical study, female subjects received a single oral dose of venetoclax 100 mg followed by a 100-µg i.v. dose of cold-labeled 13 C-venetoclax at the oral time of maximum concentration (Tmax ). The i.v. microdose was prepared as an extemporaneous, sterile compounded solution on the dosing day by pharmacists at the clinical site. Several measures were taken to ensure the sterility and safety of the i.v. preparation. A sensitive liquid chromatography-tandem mass spectrometry method was developed to allow the detection of plasma levels from the i.v. microdose. Plasma samples were collected through 72 h, and pharmacokinetic parameters were estimated using noncompartmental methods. Postdosing sample analysis demonstrated the consistency of the preparations and allowed the precise calculation of the pharmacokinetic parameters based on the actual injected dose. The absolute bioavailability of venetoclax was estimated at 5.4% under fasting conditions. Venetoclax extraction ratio was estimated to be 0.06 suggesting that the fraction transferred from the enterocytes into the liver is limiting venetoclax bioavailability. The proposed framework can be applied to other highly hydrophobic, poorly water-soluble compounds that exhibit high nonspecific binding to support the understanding of their absorption and disposition mechanisms and guide formulation development.


Subject(s)
Biological Availability , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Adult , Biomedical Research , Bridged Bicyclo Compounds, Heterocyclic/blood , Dose-Response Relationship, Drug , Drug Development , Female , Humans , Middle Aged , Sulfonamides/blood
2.
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
3.
Nanotoxicology ; 12(9): 957-974, 2018 11.
Article in English | MEDLINE | ID: mdl-30265172

ABSTRACT

The objective of our work was to investigate the effects of different types of nanoparticles on endothelial (HUVEC) and monocytic cell functions. We prepared and tested 14 different nanosystems comprising liposomes, lipid nanoparticles, polymer, and iron oxide nanoparticles. Some of the tested nanosystems contained targeting, therapeutic, or contrast agent(s). The effect of particles (0-400 µg/mL) on endothelial-monocytic cell interactions in response to TNF-α was investigated using an arterial bifurcation model and dynamic monocyte adhesion assay. Spontaneous HUVEC migration (0-100 µg/mL nanoparticles) and chemotaxis of monocytic cells towards MCP-1 in presence of particles (0-400 µg/mL) were determined using a barrier assay and a modified Boyden chamber assay, respectively. Lipid nanoparticles dose-dependently reduced monocytic cell chemotaxis and adhesion to activated HUVECs. Liposomal nanoparticles had little effect on cell migration, but one formulation induced monocytic cell recruitment by HUVECs under non-uniform shear stress by about 50%. Fucoidan-coated polymer nanoparticles (25-50 µg/mL) inhibited HUVEC migration and monocytic cell chemotaxis, and had a suppressive effect on monocytic cell recruitment under non-uniform shear stress. No significant effects of iron oxide nanoparticles on monocytic cell recruitment were observed except lauric acid and human albumin-coated particles which increased endothelial-monocytic interactions by 60-70%. Some of the iron oxide nanoparticles inhibited HUVEC migration and monocytic cell chemotaxis. These nanoparticle-induced effects are of importance for vascular cell biology and function and must be considered before the potential clinical use of some of the analyzed nanosystems in cardiovascular applications.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Monocytes/drug effects , Nanoparticles/chemistry , Nanoparticles/toxicity , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Culture Techniques , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemotaxis/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Humans , Monocytes/cytology , Surface Properties , THP-1 Cells
4.
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
5.
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
6.
Adv Drug Deliv Rev ; 119: 143-158, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28506745

ABSTRACT

Inflammation and angiogenesis drive the development and progression of multiple devastating diseases such as atherosclerosis, cancer, rheumatoid arthritis, and inflammatory bowel disease. Though these diseases have very different phenotypic consequences, they possess several common pathophysiological features in which monocyte recruitment, macrophage polarization, and enhanced vascular permeability play critical roles. Thus, developing rational targeting strategies tailored to the different stages of the journey of monocytes, from bone marrow to local lesions, and their extravasation from the vasculature in diseased tissues will advance nanomedicine. The integration of in vivo imaging uniquely allows studying nanoparticle kinetics, accumulation, clearance, and biological activity, at levels ranging from subcellular to an entire organism, and will shed light on the fate of intravenously administered nanomedicines. We anticipate that convergence of nanomedicines, biomedical engineering, and life sciences will help to advance clinically relevant therapeutics and diagnostic agents for patients with chronic inflammatory diseases.


Subject(s)
Inflammation/drug therapy , Nanoparticles/administration & dosage , Neovascularization, Pathologic/drug therapy , Animals , Drug Delivery Systems , Humans , Nanomedicine/methods
7.
Bioconjug Chem ; 28(5): 1413-1421, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28316241

ABSTRACT

Active targeting of nanoparticles through surface functionalization is a common strategy to enhance tumor delivery specificity. However, active targeting strategies tend to work against long polyethylene glycol's shielding effectiveness and associated favorable pharmacokinetics. To overcome these limitations, we developed a matrix metalloproteinase-2 sensitive surface-converting polyethylene glycol coating. This coating prevents nanoparticle-cell interaction in the bloodstream, but, once exposed to matrix metalloproteinase-2, i.e., when the nanoparticles accumulate within the tumor interstitium, the converting polyethylene glycol coating is cleaved, and targeting ligands become available for binding to tumor cells. In this study, we applied a comprehensive multimodal imaging strategy involving optical, nuclear, and magnetic resonance imaging methods to evaluate this coating approach in a breast tumor mouse model. The data obtained revealed that this surface-converting coating enhances the nanoparticle's blood half-life and tumor accumulation and ultimately results in improved tumor-cell targeting. Our results show that this enzyme-specific surface-converting coating ensures a high cell-targeting specificity without compromising favorable nanoparticle pharmacokinetics.


Subject(s)
Breast Neoplasms/pathology , Magnetic Resonance Imaging/methods , Matrix Metalloproteinase 2/metabolism , Multimodal Imaging/methods , Nanoparticles/administration & dosage , Spectrophotometry, Infrared/methods , Animals , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Female , Humans , Image Processing, Computer-Assisted/methods , Matrix Metalloproteinase 2/chemistry , Mice , Mice, Nude , Nanoparticles/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Angew Chem Int Ed Engl ; 56(11): 2923-2926, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28112478

ABSTRACT

Understanding the formation process of nanoparticles is of the utmost importance to improve their design and production. This especially holds true for self-assembled nanoparticles whose formation processes have been largely overlooked. Herein, we present a new technology that integrates a microfluidic-based nanoparticle synthesis method and Förster resonance energy transfer (FRET) microscopy imaging to visualize nanoparticle self-assembly in real time. Applied to different nanoparticle systems, for example, nanoemulsions, drug-loaded block-copolymer micelles, and nanocrystal-core reconstituted high-density lipoproteins, we have shown the approach's unique ability to investigate key parameters affecting nanoparticle formation.


Subject(s)
Fluorescence Resonance Energy Transfer , Nanoparticles/chemistry , Time Factors
9.
Proc Natl Acad Sci U S A ; 113(44): E6731-E6740, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791119

ABSTRACT

Immunological complexity in atherosclerosis warrants targeted treatment of specific inflammatory cells that aggravate the disease. With the initiation of large phase III trials investigating immunomodulatory drugs for atherosclerosis, cardiovascular disease treatment enters a new era. We here propose a radically different approach: implementing and evaluating in vivo a combinatorial library of nanoparticles with distinct physiochemical properties and differential immune cell specificities. The library's nanoparticles are based on endogenous high-density lipoprotein, which can preferentially deliver therapeutic compounds to pathological macrophages in atherosclerosis. Using the apolipoprotein E-deficient (Apoe-/-) mouse model of atherosclerosis, we quantitatively evaluated the library's immune cell specificity by combining immunological techniques and in vivo positron emission tomography imaging. Based on this screen, we formulated a liver X receptor agonist (GW3965) and abolished its liver toxicity while still preserving its therapeutic function. Screening the immune cell specificity of nanoparticles can be used to develop tailored therapies for atherosclerosis and other inflammatory diseases.


Subject(s)
Atherosclerosis/drug therapy , Atherosclerosis/immunology , Immunotherapy , Nanoparticles/chemistry , Animals , Anti-Inflammatory Agents , Apolipoproteins E/deficiency , Atherosclerosis/pathology , Autoradiography , Benzoates/agonists , Benzoates/chemistry , Benzylamines/agonists , Benzylamines/chemistry , Disease Models, Animal , Drug Delivery Systems/methods , Female , Gene Expression Regulation/drug effects , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Imaging , Nanomedicine , Nanoparticles/metabolism , Positron-Emission Tomography/methods , RNA, Messenger/metabolism
10.
Int J Nanomedicine ; 11: 5027-5040, 2016.
Article in English | MEDLINE | ID: mdl-27785012

ABSTRACT

Inflammation, oxidative stress, and uncontrolled cell proliferation are common key features of chronic inflammatory diseases, such as atherosclerosis and cancer. ω3 polyunsaturated fatty acids (PUFAs; also known as omega3 fatty acids or fish oil) have beneficial effects against inflammation upon dietary consumption. However, these effects cannot be fully exploited unless diets are enriched with high concentrations of fish oil supplements over long periods of time. Here, a nanomedicine-based approach is presented for delivering effective levels of PUFAs to inflammatory cells. Nanoparticles are internalized by immune cells, and hence can adequately deliver bioactive lipids into these target cells. The ω3 FA docosahexaenoic acid was formulated into liposomes (ω-liposomes), and evaluated for anti-inflammatory effects in different types of immune cells. ω-Liposomes strongly inhibited the release of reactive oxygen species and reactive nitrogen species from human neutrophils and murine macrophages, and also inhibited the production of the proinflammatory cytokines TNFα and MCP1. Moreover, ω-liposomes inhibited tumor-cell proliferation when evaluated in FaDu head and neck squamous carcinoma and 4T1 breast cancer cells in in vitro cultures. We propose that ω-liposomes are a promising nanonutraceutical formulation for intravenous delivery of fish oil FAs, which may be beneficial in the treatment of inflammatory disorders and cancer.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Docosahexaenoic Acids/pharmacology , Inflammation/drug therapy , Liposomes/administration & dosage , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Cytokines/metabolism , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , In Vitro Techniques , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice , Monocytes/drug effects , Monocytes/metabolism , Oxidative Stress/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Vascul Pharmacol ; 82: 51-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27189780

ABSTRACT

BACKGROUND: Macrophages play a central role in atherosclerosis development and progression, hence, targeting macrophage activity is considered an attractive therapeutic. Recently, we documented nanomedicinal delivery of the anti-inflammatory compound prednisolone to atherosclerotic plaque macrophages in patients, which did however not translate into therapeutic efficacy. This unanticipated finding calls for in-depth screening of drugs intended for targeting plaque macrophages. METHODS AND RESULTS: We evaluated the effect of several candidate drugs on macrophage activity, rating overall performance with respect to changes in cytokine release, oxidative stress, lipid handling, endoplasmic reticulum (ER) stress, and proliferation of macrophages. Using this in vitro approach, we observed that the anti-inflammatory effect of prednisolone was counterbalanced by multiple adverse effects on other key pathways. Conversely, pterostilbene, T0901317 and simvastatin had an overall anti-atherogenic effect on multiple pathways, suggesting their potential for liposomal delivery. CONCLUSION: This dedicated assay setup provides a framework for high-throughput assessment. Further in vivo studies are warranted to determine the predictive value of this macrophage-based screening approach and its potential value in nanomedicinal drug development for cardiovascular patients.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atherosclerosis/drug therapy , Inflammation/drug therapy , Macrophage Activation/drug effects , Macrophages/drug effects , Plaque, Atherosclerotic , Signal Transduction/drug effects , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Proliferation/drug effects , Cytokines/metabolism , Endoplasmic Reticulum Stress/drug effects , High-Throughput Screening Assays , Humans , Hydrocarbons, Fluorinated/pharmacology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Lipid Metabolism/drug effects , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress/drug effects , Prednisolone/pharmacology , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Simvastatin/pharmacology , Stilbenes/pharmacology , Sulfonamides/pharmacology , Transfection
12.
Nanomedicine ; 12(6): 1463-70, 2016 08.
Article in English | MEDLINE | ID: mdl-27015770

ABSTRACT

Atherosclerosis is a lipid-driven inflammatory disease, for which nanomedicinal interventions are under evaluation. Previously, we showed that liposomal nanoparticles loaded with prednisolone (LN-PLP) accumulated in plaque macrophages, however, induced proatherogenic effects in patients. Here, we confirmed in low-density lipoprotein receptor knockout (LDLr(-/-)) mice that LN-PLP accumulates in plaque macrophages. Next, we found that LN-PLP infusions at 10mg/kg for 2weeks enhanced monocyte recruitment to plaques. In follow up, after 6weeks of LN-PLP exposure we observed (i) increased macrophage content, (ii) more advanced plaque stages, and (iii) larger necrotic core sizes. Finally, in vitro studies showed that macrophages become lipotoxic after LN-PLP exposure, exemplified by enhanced lipid loading, ER stress and apoptosis. These findings indicate that liposomal prednisolone may paradoxically accelerate atherosclerosis by promoting macrophage lipotoxicity. Hence, future (nanomedicinal) drug development studies are challenged by the multifactorial nature of atherosclerotic inflammation.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Prednisolone/administration & dosage , Animals , Humans , Liposomes , Macrophages/pathology , Mice , Plaque, Atherosclerotic
13.
Sci Rep ; 6: 22519, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26931825

ABSTRACT

Cells release nano-sized membrane vesicles that are involved in intercellular communication by transferring biological information between cells. It is generally accepted that cells release at least three types of extracellular vesicles (EVs): apoptotic bodies, microvesicles and exosomes. While a wide range of putative biological functions have been attributed to exosomes, they are assumed to represent a homogenous population of EVs. We hypothesized the existence of subpopulations of exosomes with defined molecular compositions and biological properties. Density gradient centrifugation of isolated exosomes revealed the presence of two distinct subpopulations, differing in biophysical properties and their proteomic and RNA repertoires. Interestingly, the subpopulations mediated differential effects on the gene expression programmes in recipient cells. In conclusion, we demonstrate that cells release distinct exosome subpopulations with unique compositions that elicit differential effects on recipient cells. Further dissection of exosome heterogeneity will advance our understanding of exosomal biology in health and disease and accelerate the development of exosome-based diagnostics and therapeutics.


Subject(s)
Exosomes/metabolism , Melanoma, Experimental/pathology , Animals , Cell Line, Tumor , Mice
14.
Front Physiol ; 4: 365, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24379786

ABSTRACT

Hereditary hemolytic anemia encompasses a heterogeneous group of anemias characterized by decreased red blood cell survival because of inherited membrane, enzyme, or hemoglobin disorders. Affected red blood cells are more fragile, less deformable, and more susceptible to shear stress and oxidative damage, and show increased vesiculation. Red blood cells, as essentially all cells, constitutively release phospholipid extracellular vesicles in vivo and in vitro in a process known as vesiculation. These extracellular vesicles comprise a heterogeneous group of vesicles of different sizes and intracellular origins. They are described in literature as exosomes if they originate from multi-vesicular bodies, or as microvesicles when formed by a one-step budding process directly from the plasma membrane. Extracellular vesicles contain a multitude of bioactive molecules that are implicated in intercellular communication and in different biological and pathophysiological processes. Mature red blood cells release in principle only microvesicles. In hereditary hemolytic anemias, the underlying molecular defect affects and determines red blood cell vesiculation, resulting in shedding microvesicles of different compositions and concentrations. Despite extensive research into red blood cell biochemistry and physiology, little is known about red cell deformability and vesiculation in hereditary hemolytic anemias, and the associated pathophysiological role is incompletely assessed. In this review, we discuss recent progress in understanding extracellular vesicles biology, with focus on red blood cell vesiculation. Also, we review recent scientific findings on the molecular defects of hereditary hemolytic anemias, and their correlation with red blood cell deformability and vesiculation. Integrating bio-analytical findings on abnormalities of red blood cells and their microvesicles will be critical for a better understanding of the pathophysiology of hereditary hemolytic anemias.

SELECTION OF CITATIONS
SEARCH DETAIL
...