Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38293207

ABSTRACT

Ischemic stroke-induced mitochondrial dysfunction in the blood-brain barrier-forming brain endothelial cells ( BECs ) results in long-term neurological dysfunction post-stroke. We previously reported that intravenous administration of human BEC ( hBEC )-derived mitochondria-containing extracellular vesicles ( EVs ) showed a potential efficacy signal in a mouse middle cerebral artery occlusion ( MCAo ) model of stroke. We hypothesized that EVs harvested from donor species homologous to the recipient species ( e.g., mouse) may improve therapeutic efficacy, and therefore, use of mouse BEC ( mBEC )-derived EVs may improve post-stroke outcomes in MCAo mice. We investigated if EVs derived from the same species as the recipient cell (mBEC-EVs and recipient mBECs or hBECs-EVs and recipient hBECs) show a greater EV mitochondria delivery efficiency than cross-species EVs and recipient cells (mBEC-EVs and recipient hBECs or vice versa ). Our results showed that mBEC-EVs outperformed hBEC-EVs in transferring EV mitochondria to the recipient ischemic mBECs, and improved mBEC mitochondrial function via increasing oxygen consumption rate. mBEC-EVs significantly reduced brain infarct volume and improved behavioral recovery compared to vehicle-injected MCAo mice. Our data suggests that mBEC-EVs show superior therapeutic efficacy in a mouse MCAo stroke model compared to hBEC-EVs-supporting the continued use of mBEC-EVs to optimize the therapeutic potential of mitochondria-containing EVs in preclinical studies.

2.
J Control Release ; 365: 801, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38043728
3.
Expert Opin Drug Deliv ; 20(12): 1769-1788, 2023.
Article in English | MEDLINE | ID: mdl-37921194

ABSTRACT

INTRODUCTION: Ischemic stroke-induced mitochondrial dysfunction in brain endothelial cells (BECs) leads to breakdown of the blood-brain barrier (BBB) causing long-term neurological dysfunction. Restoration of mitochondrial function in injured BECs is a promising therapeutic strategy to alleviate stroke-induced damage. Mounting evidence demonstrate that selected subsets of cell-derived extracellular vehicles (EVs), such as exosomes (EXOs) and microvesicles (MVs), contain functional mitochondrial components. Therefore, development of BEC-derived mitochondria-containing EVs for delivery to the BBB will (1) alleviate mitochondrial dysfunction and limit long-term neurological dysfunction in ischemic stroke and (2) provide an alternative therapeutic option for treating numerous other diseases associated with mitochondrial dysfunction. AREA COVERED: This review will discuss (1) how EV subsets package different types of mitochondrial components during their biogenesis, (2) mechanisms of EV internalization and functional mitochondrial responses in the recipient cells, and (3) EV biodistribution and pharmacokinetics - key factors involved in the development of mitochondria-containing EVs as a novel BBB-targeted stroke therapy. EXPERT OPINION: Mitochondria-containing MVs have demonstrated therapeutic benefits in ischemic stroke and other pathologies associated with mitochondrial dysfunction. Delivery of MV mitochondria to the BBB is expected to protect the BBB integrity and neurovascular unit post-stroke. MV mitochondria quality control, characterization, mechanistic understanding of its effects in vivo, safety and efficacy in different preclinical models, large-scale production, and establishment of regulatory guidelines are foreseeable milestones to harness the clinical potential of MV mitochondria delivery.


Subject(s)
Extracellular Vesicles , Ischemic Stroke , Mitochondrial Diseases , Stroke , Humans , Blood-Brain Barrier/metabolism , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Endothelial Cells/metabolism , Tissue Distribution , Extracellular Vesicles/metabolism , Stroke/drug therapy , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology
4.
J Control Release ; 354: 368-393, 2023 02.
Article in English | MEDLINE | ID: mdl-36642252

ABSTRACT

Ischemic stroke causes brain endothelial cell (BEC) death and damages tight junction integrity of the blood-brain barrier (BBB). We harnessed the innate mitochondrial load of BEC-derived extracellular vesicles (EVs) and utilized mixtures of EV/exogenous 27 kDa heat shock protein (HSP27) as a one-two punch strategy to increase BEC survival (via EV mitochondria) and preserve their tight junction integrity (via HSP27 effects). We demonstrated that the medium-to-large (m/lEV) but not small EVs (sEV) transferred their mitochondrial load, that subsequently colocalized with the mitochondrial network of the recipient primary human BECs. Recipient BECs treated with m/lEVs showed increased relative ATP levels and mitochondrial function. To determine if the m/lEV-meditated increase in recipient BEC ATP levels was associated with m/lEV mitochondria, we isolated m/lEVs from donor BECs pre-treated with oligomycin A (OGM, mitochondria electron transport complex V inhibitor), referred to as OGM-m/lEVs. BECs treated with naïve m/lEVs showed a significant increase in ATP levels compared to untreated OGD cells, OGM-m/lEVs treated BECs showed a loss of ATP levels suggesting that the m/lEV-mediated increase in ATP levels is likely a function of their innate mitochondrial load. In contrast, sEV-mediated ATP increases were not affected by inhibition of mitochondrial function in the donor BECs. Intravenously administered m/lEVs showed a reduction in brain infarct sizes compared to vehicle-injected mice in a mouse middle cerebral artery occlusion model of ischemic stroke. We formulated binary mixtures of human recombinant HSP27 protein with EVs: EV/HSP27 and ternary mixtures of HSP27 and EVs with a cationic polymer, poly (ethylene glycol)-b-poly (diethyltriamine): (PEG-DET/HSP27)/EV. (PEG-DET/HSP27)/EV and EV/HSP27 mixtures decreased the paracellular permeability of small and large molecular mass fluorescent tracers in oxygen glucose-deprived primary human BECs. This one-two punch approach to increase BEC metabolic function and tight junction integrity may be a promising strategy for BBB protection and prevention of long-term neurological dysfunction post-ischemic stroke.


Subject(s)
Extracellular Vesicles , Ischemic Stroke , Stroke , Mice , Humans , Animals , HSP27 Heat-Shock Proteins/metabolism , Brain/metabolism , Blood-Brain Barrier/metabolism , Stroke/metabolism , Infarction, Middle Cerebral Artery/metabolism , Heat-Shock Proteins/metabolism , Ischemic Stroke/metabolism , Mitochondria/metabolism , Extracellular Vesicles/metabolism , Adenosine Triphosphate/metabolism
5.
Cell Mol Bioeng ; 15(5): 367-389, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36444353

ABSTRACT

Introduction: Extracellular vesicles (EVs) are promising carriers for the delivery of biotherapeutic cargo such as RNA and proteins. We have previously demonstrated that the innate EV mitochondria in microvesicles (MVs), but not exosomes (EXOs) can be transferred to recipient BECs and mouse brain slice neurons. Here, we sought to determine if the innate EV mitochondrial load can be further increased via increasing mitochondrial biogenesis in the donor cells. We hypothesized that mitochondria-enriched EVs ("mito-EVs") may increase the recipient BEC ATP levels to a greater extent than naïve MVs. Methods: We treated NIH/3T3, a fibroblast cell line and hCMEC/D3, a human brain endothelial cell (BEC) line using resveratrol to activate peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), the central mediator of mitochondrial biogenesis. Naïve EVs and mito-EVs isolated from the non-activated and activated donor cells were characterized using transmission electron microscopy, dynamic light scattering and nanoparticle tracking analysis. The effect of mito-EVs on resulting ATP levels in the recipient BECs were determined using Cell Titer Glo ATP assay. The uptake of Mitotracker Red-stained EVs into recipient BECs and their colocalization with recipient BEC mitochondria were studied using flow cytometry and fluorescence microscopy. Results: Resveratrol treatment increased PGC-1α expression in the donor cells. Mito-MVs but not mito-EXOs showed increased expression of mitochondrial markers ATP5A and TOMM20 compared to naïve MVs. TEM images showed that a greater number of mito-MVs contained mitochondria compared to naïve MVs. Mito-MVs but not mito-EXOs showed a larger particle diameter compared to their naïve EV counterparts from the non-activated cells suggesting increased mitochondria incorporation. Mito-EVs were generated at higher particle concentrations compared to naïve EVs from non-activated cells. Mito-EVs increased the cellular ATP levels and transferred their mitochondrial load into the recipient BECs. Mito-MV mitochondria also colocalized with recipient BEC mitochondria. Conclusions: Our results suggest that the pharmacological modulation of mitochondrial biogenesis in the donor cells can change the mitochondrial load in the secreted MVs. Outcomes of physicochemical characterization studies and biological assays confirmed the superior effects of mito-MVs compared to naïve MVs-suggesting their potential to improve mitochondrial function in neurovascular and neurodegenerative diseases. Supplementary Information: The online version contains supplementary material available at 10.1007/s12195-022-00738-8.

6.
J Control Release ; 343: 400-407, 2022 03.
Article in English | MEDLINE | ID: mdl-35131369

ABSTRACT

The field of drug delivery has made tremendous advances in increasing the therapeutic potential of a variety of drug candidates spanning from small molecules to large molecular biologics such as nucleic acids, proteins, etc. Extracellular vesicles (EVs) are mediators of intercellular communication and carry a rich cocktail of innate cargo including lipids, proteins and nucleic acids. EVs are a promising class of natural, cell-derived carriers for drug delivery. EVs of particle diameters <200 nm are referred to as small EVs (sEVs) and medium-to-larger particles of diameters >200 nm are referred to as m/lEVs. The m/lEVs naturally incorporate mitochondria during their biogenesis. In this Oration, I will discuss the potential of m/lEVs as carriers for the delivery of healthy and functional mitochondria. Mitochondrial damage and dysfunction play a causal role in multiple pathologies such as neurodegenerative diseases, cardiovascular and metabolic diseases-suggesting that m/lEV-mediated mitochondria delivery can be of broad biomedical significance. A major advantage of harnessing m/lEVs is that the delivered mitochondria are capable of using endogenous mechanisms for repairing the cellular damage. I will highlight the delivery potential of m/lEVs based on the studies we have conducted so far and discuss unaddressed issues towards their development as a novel class of mitochondria carriers.


Subject(s)
Extracellular Vesicles , Nucleic Acids , Drug Delivery Systems , Extracellular Vesicles/metabolism , Mitochondria/metabolism , Proteins/metabolism
7.
AAPS J ; 24(1): 8, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34873640

ABSTRACT

Lipidoid nanoparticles (LNPs) are the delivery platform in Onpattro, the first FDA-approved siRNA drug. LNPs are also the carriers in the Pfizer-BioNTech and Moderna COVID-19 mRNA vaccines. While these applications have demonstrated that LNPs effectively deliver nucleic acids to hepatic and muscle cells, it is unclear if LNPs could be used for delivery of siRNA to neural cells, which are notoriously challenging delivery targets. Therefore, the purpose of this study was to determine if LNPs could efficiently deliver siRNA to neurons. Because of their potential delivery utility in either applications for the central nervous system and the peripheral nervous system, we used both cortical neurons and sensory neurons. We prepared siRNA-LNPs using C12-200, a benchmark ionizable cationic lipidoid along with helper lipids. We demonstrated using dynamic light scattering that the inclusion of both siRNA and PEG-lipid provided a stabilizing effect to the LNP particle diameters and polydispersity indices by minimizing aggregation. We found that siRNA-LNPs were safely tolerated by primary dorsal root ganglion neurons. Flow cytometry analysis revealed that Cy5 siRNA delivered via LNPs into rat primary cortical neurons showed uptake levels similar to Lipofectamine RNAiMAX-the gold standard commercial transfection agent. However, LNPs demonstrated a superior safety profile, whereas the Lipofectamine-mediated uptake was concomitant with significant toxicity. Fluorescence microscopy demonstrated a time-dependent increase in the uptake of LNP-delivered Cy5 siRNA in a human cortical neuron cell line. Overall, our results suggest that LNPs are a viable platform that can be optimized for delivery of therapeutic siRNAs to neural cells.


Subject(s)
Ganglia, Spinal/metabolism , Lipids/chemistry , Nanoparticles , Neurons/metabolism , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Transfection , Animals , Carbocyanines/metabolism , Fluorescent Dyes/metabolism , Ganglia, Spinal/cytology , Humans , MCF-7 Cells , Microscopy, Fluorescence , Nanotechnology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Time Factors
8.
J Control Release ; 338: 505-526, 2021 10 10.
Article in English | MEDLINE | ID: mdl-34450196

ABSTRACT

We have demonstrated, for the first time that microvesicles, a sub-type of extracellular vesicles (EVs) derived from hCMEC/D3: a human brain endothelial cell (BEC) line transfer polarized mitochondria to recipient BECs in culture and to neurons in mice acute brain cortical and hippocampal slices. This mitochondrial transfer increased ATP levels by 100 to 200-fold (relative to untreated cells) in the recipient BECs exposed to oxygen-glucose deprivation, an in vitro model of cerebral ischemia. We have also demonstrated that transfer of microvesicles, the larger EV fraction, but not exosomes resulted in increased mitochondrial function in hypoxic endothelial cultures. Gene ontology and pathway enrichment analysis of EVs revealed a very high association to glycolysis-related processes. In comparison to heterotypic macrophage-derived EVs, BEC-derived EVs demonstrated a greater selectivity to transfer mitochondria and increase endothelial cell survival under ischemic conditions.


Subject(s)
Cell-Derived Microparticles , Extracellular Vesicles , Animals , Brain , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Mice , Mitochondria
9.
PLoS One ; 15(4): e0231597, 2020.
Article in English | MEDLINE | ID: mdl-32287325

ABSTRACT

Resident microglia of the central nervous system are being increasingly recognized as key players in diseases such as neuropathic pain. Biochemical and behavioral studies in neuropathic pain rodent models have documented compelling evidence of the critical role of ATP mediated-P2X4R-brain-derived neurotrophic factor (BDNF) signaling pathway in the initiation and maintenance of pain hypersensitivity, a feature driving neuropathic pain-related behavior. The goal of this study was to develop and characterize an in vitro cell line model of activated microglia that can be subsequently utilized for screening neuropathic pain therapeutics. In the present study, we characterized the SIM-A9 microglia cell line for key molecules in the P2X4R-BDNF signaling axis using a combination of biochemical techniques and developed an ATP-activated SIM-A9 microglia model. We present three novel findings: first, SIM-A9 cells expressed P2X4R and BDNF proteins, second, ATP, but not LPS, was cytocompatible with SIM-A9 cells and third, exposure of cells to optimized ATP concentrations for defined periods increased intracellular expression of Iba1 and BDNF proteins. Increased Iba1 levels confirmed microglia activation and increased BDNF expression confirmed ATP-mediated stimulation of the P2X4R signaling pathway. We propose that this ATP-activated SIM-A9 cell line model system can be utilized for screening both small- as well as macro-molecular neuropathic pain therapeutics targeting BDNF and/or P2X4R knockdown.


Subject(s)
Microglia/metabolism , Neuralgia/metabolism , Signal Transduction , Adenosine Triphosphate/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Culture Techniques/methods , Lipopolysaccharides/pharmacology , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/cytology , Microglia/drug effects , Neuralgia/pathology , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism
10.
J Vis Exp ; (150)2019 08 19.
Article in English | MEDLINE | ID: mdl-31475964

ABSTRACT

The blood-brain barrier BBB consists of endothelial cells that form a barrier between the systemic circulation and the brain to prevent the exchange of non-essential ions and toxic substances. Tight junctions (TJ) effectively seal the paracellular space in the monolayers resulting in an intact barrier. This study describes a LY-based fluorescence assay that can be used to determine its apparent permeability coefficient (Papp) and in turn can be used to determine the kinetics of the formation of confluent monolayers and the resulting tight junction barrier integrity in hCMEC/D3 monolayers. We further demonstrate an additional utility of this assay to determine TJ functional integrity in transfected cells. Our data from the LY Papp assay shows that the hCMEC/D3 cells seeded in a transwell setup effectively limit LY paracellular transport 7 days-post culture. As an additional utility of the presented assay, we also demonstrate that the DNA nanoparticle transfection does not alter LY paracellular transport in hCMEC/D3 monolayers.


Subject(s)
Blood-Brain Barrier , Capillary Permeability , Isoquinolines/metabolism , Biological Transport , Biomarkers , Blood-Brain Barrier/cytology , Brain , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Models, Neurological , Tight Junctions/metabolism
11.
Adv Funct Mater ; 28(6)2018 Feb 07.
Article in English | MEDLINE | ID: mdl-29785179

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is identified as a potent neuroprotective and neuroregenerative agent for many neurological diseases. Regrettably, its delivery to the brain is hampered by poor serum stability and rapid brain clearance. Here, a novel nanoformulation is reported composed of a bio-compatible polymer, poly(ethylene glycol)-b-poly(L-glutamic acid) (PEG-PLE), that hosts the BDNF molecule in a nanoscale complex, termed here Nano-BDNF. Upon simple mixture, Nano-BDNF spontaneously forms uniform spherical particles with a core-shell structure. Molecular dynamics simulations suggest that binding between BDNF and PEG-PLE is mediated through electrostatic coupling as well as transient hydrogen bonding. The formation of Nano-BDNF complex stabilizes BDNF and protects it from nonspecific binding with common proteins in the body fluid, while allowing it to associate with its receptors. Following intranasal administration, the nanoformulation improves BDNF delivery throughout the brain and displays a more preferable regional distribution pattern than the native protein. Furthermore, intranasally delivered Nano-BDNF results in superior neuroprotective effects in the mouse brain with lipopolysaccharides-induced inflammation, indicating promise for further evaluation of this agent for the therapy of neurologic diseases.

12.
Pharmacol Biochem Behav ; 150-151: 48-56, 2016.
Article in English | MEDLINE | ID: mdl-27619636

ABSTRACT

BACKGROUND: Low levels of brain-derived neurotrophic factor (BDNF) are linked to delayed neurological recovery, depression, and cognitive impairment following stroke. Supplementation with BDNF reverses these effects. Unfortunately, systemically administered BDNF in its native form has minimal therapeutic value due to its poor blood brain barrier permeability and short serum half-life. In this study, a novel nano-particle polyion complex formulation of BDNF (nano-BDNF) was administered to mice after experimental ischemic stroke. METHODS: Male C57BL/6J (8-10weeks) mice were randomly assigned to receive nano-BDNF, native-BDNF, or saline treatment after being subjected to 60min of reversible middle cerebral artery occlusion (MCAo). Mice received the first dose at 3 (early treatment), 6 (intermediate treatment), or 12h (delayed treatment) following stroke onset; a second dose was given in all cohorts at 24h after stroke onset. Post-stroke outcome was evaluated by behavioral, histological, and molecular analysis for 15days after stroke. RESULTS: Early and intermediate nano-BDNF treatment led to a significant reduction in cerebral tissue loss. Delayed treatment led to improved memory/cognition, reduced post-stroke depressive phenotypes, and maintained myelin basic protein and brain BDNF levels, but had no effect on tissue atrophy. CONCLUSIONS: The results indicate that administration of a novel nano-particle formulation of BDNF leads to both neuroprotective and neuro-restorative effects after stroke.


Subject(s)
Behavior, Animal/drug effects , Brain Ischemia/drug therapy , Brain-Derived Neurotrophic Factor/administration & dosage , Nanoparticles/administration & dosage , Animals , Cyclic AMP Response Element-Binding Protein/analysis , Depression/drug therapy , Drug Compounding , Drug Delivery Systems , Male , Memory/drug effects , Mice , Mice, Inbred C57BL
13.
J Control Release ; 231: 38-49, 2016 06 10.
Article in English | MEDLINE | ID: mdl-26928528

ABSTRACT

We previously developed a "cage"-like nano-formulation (nanozyme) for copper/Zinc superoxide dismutase (SOD1) by polyion condensation with a conventional block copolymer poly(ethylene glycol)-b-poly(L-lysine) (PEG-PLL) followed by chemical cross-linking. Herein we report a new SOD1 nanozyme based on PEG-b-poly(aspartate diethyltriamine) (PEG-PAsp(DET), or PEG-DET for short) engineered for chronic dosing. This new nanozyme was spherical (Rg/Rh=0.785), and hollow (60% water composition) nanoparticles with colloidal properties similar to PLL-based nanozyme. It was better tolerated by brain microvessel endothelial/neuronal cells, and accumulated less in the liver and spleen. This formulation reduced the infarct volumes by more than 50% in a mouse model of ischemic stroke. However, it was not effective at preventing neuromuscular junction denervation in a mutant SOD1(G93A) mouse model of amyotrophic lateral sclerosis (ALS). To our knowledge, this work is the first report of using PEG-DET for protein delivery and a direct comparison between two cationic block copolymers demonstrating the effect of polymer structure in modulating the mononuclear phagocyte system (MPS) accumulation of polyion complexes.


Subject(s)
Antioxidants/pharmacology , Mononuclear Phagocyte System/drug effects , Nanoparticles/chemistry , Superoxide Dismutase-1/pharmacology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Antioxidants/chemistry , Antioxidants/toxicity , Brain/blood supply , Brain/drug effects , Brain/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/drug effects , Microvessels/pathology , Mononuclear Phagocyte System/pathology , Mutation , Neurons/drug effects , Neurons/pathology , Polyethylene Glycols/chemistry , Proteins/chemistry , Stroke/drug therapy , Stroke/pathology , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/toxicity , Tissue Distribution
14.
Obesity (Silver Spring) ; 24(1): 148-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26612356

ABSTRACT

OBJECTIVE: An intimate association exists between oxidative stress and inflammation. Because adipose tissue (AT) inflammation is intricately linked to metabolic disorders, it was hypothesized that reducing oxidative stress would be effective in ameliorating AT inflammation in obesity. METHODS: Wild-type mice were fed a high-fat diet (HF) for 8 weeks followed by a 2-week treatment with nanoformulated copper/zinc superoxide dismutase (NanoSOD). The mice were divided into: 1) chow diet, 2) HF, and 3) HF + NanoSOD. RESULTS: The HF + NanoSOD-treated mice showed a significant decrease in plasma and liver triglycerides when compared with HF-fed mice. Interestingly, NanoSOD reduced the expression of macrophage and inflammatory markers in visceral AT (VAT) and stromal cells derived from VAT. Moreover, the activation of proinflammatory signaling pathways, in particular, the extracellular signal-regulated kinases, was blunted in VAT on NanoSOD treatment. However, markers of oxidative stress were not altered significantly in the HF + NanoSOD group in the experimental conditions. Pretreatment of either macrophages or adipocytes significantly reduced the inflammatory response invoked in an in vitro coculture system, further supporting the role of NanoSOD in inhibiting obesity-linked inflammation. CONCLUSIONS: This data suggest that NanoSOD is effective not only in reducing AT macrophage accumulation and AT inflammation but also in promoting triglyceride metabolism in obesity.


Subject(s)
Adipose Tissue/drug effects , Inflammation/prevention & control , Obesity/pathology , Superoxide Dismutase/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adiposity/drug effects , Animals , Diet, High-Fat , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation/metabolism , Lipid Metabolism/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Obesity/complications , Obesity/metabolism , Superoxide Dismutase/administration & dosage , Triglycerides/blood
15.
Biochem Biophys Res Commun ; 469(3): 495-500, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26692492

ABSTRACT

OBJECTIVE: Endothelial cell (EC) oxidative stress can lead to vascular dysfunction which is an underlying event in the development of cardiovascular disease (CVD). The lack of a potent and bioavailable anti-oxidant enzyme is a major challenge in studies on antioxidant therapy. The objective of this study is to determine whether copper/zinc superoxide dismutase (CuZnSOD or SOD1) after nanoformulation (nanoSOD) can effectively reduce EC oxidative stress and/or vascular inflammation in obesity. METHODS: Human aortic endothelial cells (HAECs) were treated with native- or nanoSOD for 6 h followed by treatment with linoleic acid (LA), a free fatty acid, for 6-24 h. To determine the in vivo relevance, the effectiveness of nanoSOD in reducing vascular cell activation was studied in a mouse model of diet-induced obesity. RESULTS: We noted that nanoSOD was more effectively taken up by ECs than native SOD. Western blot analysis further confirmed that the intracellular accumulation of SOD1 protein was greatly increased upon nanoSOD treatment. Importantly, nanoSOD pretreatment led to a significant decrease in LA-induced oxidative stress in ECs which was associated with a marked increase in SOD enzyme activity in ECs. In vivo studies showed a significant decrease in markers of EC/vascular cell activation and/or inflammation in visceral adipose tissue (VAT), thoracic aorta, and heart collected from nanoSOD-treated mice compared to obese control mice. Interestingly, the expression of metallothionein 2, an antioxidant gene was significantly increased in nanoSOD-treated mice. CONCLUSION: Our data show that nanoSOD is very effective in delivering active SOD to ECs and in reducing EC oxidative stress. Our data also demonstrate that nanoSOD will be a useful tool to reduce vascular cell activation in VAT and aorta in obesity which, in turn, can protect against obesity-associated CVD, in particular, hypertension.


Subject(s)
Aortitis/drug therapy , Aortitis/immunology , Endothelial Cells/immunology , Obesity/drug therapy , Obesity/immunology , Superoxide Dismutase/administration & dosage , Animals , Cells, Cultured , Drug Compounding , Endothelial Cells/drug effects , Free Radical Scavengers/administration & dosage , Humans , Mice , Mice, Inbred C57BL , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Treatment Outcome
16.
Oxid Med Cell Longev ; 2015: 5194239, 2015.
Article in English | MEDLINE | ID: mdl-26697135

ABSTRACT

Use of antioxidants to mitigate oxidative stress during ocular inflammatory diseases has shown therapeutic potential. This work examines a nanoscale therapeutic modality for the eye on the base of antioxidant enzyme, superoxide dismutase 1 (SOD1), termed "nanozyme." The nanozyme is produced by electrostatic coupling of the SOD1 with a cationic block copolymer, poly(L-lysine)-poly(ethyleneglycol), followed by covalent cross-linking of the complexes with 3,3'-dithiobis(sulfosuccinimidylpropionate) sodium salt. The ability of SOD1 nanozyme as well as the native SOD1 to reduce inflammatory processes in the eye was examined in vivo in rabbits with immunogenic uveitis. Results suggested that topical instillations of both enzyme forms demonstrated anti-inflammatory activity; however, the nanozyme was much more effective compared to the free enzyme in decreasing uveitis manifestations. In particular, we noted statistically significant differences in such inflammatory signs in the eye as the intensities of corneal and iris edema, hyperemia of conjunctiva, lens opacity, fibrin clots, and the protein content in aqueous humor. Clinical findings were confirmed by histological data. Thus, SOD1-containing nanozyme is potentially useful therapeutic agent for the treatment of ocular inflammatory disorders.


Subject(s)
Superoxide Dismutase/therapeutic use , Uveitis/drug therapy , Animals , Conjunctiva/metabolism , Conjunctiva/pathology , Polymers/chemistry , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Succinimides/chemistry , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Uveitis/metabolism , Uveitis/pathology
17.
J Control Release ; 190: 637-63, 2014 Sep 28.
Article in English | MEDLINE | ID: mdl-24956489

ABSTRACT

A variety of therapeutic proteins have shown potential to treat central nervous system (CNS) disorders. Challenge to deliver these protein molecules to the brain is well known. Proteins administered through parenteral routes are often excluded from the brain because of their poor bioavailability and the existence of the blood-brain barrier (BBB). Barriers also exist to proteins administered through non-parenteral routes that bypass the BBB. Several strategies have shown promise in delivering proteins to the brain. This review, first, describes the physiology and pathology of the BBB that underscore the rationale and needs of each strategy to be applied. Second, major classes of protein therapeutics along with some key factors that affect their delivery outcomes are presented. Third, different routes of protein administration (parenteral, central intracerebroventricular and intraparenchymal, intranasal and intrathecal) are discussed along with key barriers to CNS delivery associated with each route. Finally, current delivery strategies involving chemical modification of proteins and use of particle-based carriers are overviewed using examples from literature and our own work. Whereas most of these studies are in the early stage, some provide proof of mechanism of increased protein delivery to the brain in relevant models of CNS diseases, while in few cases proof of concept had been attained in clinical studies. This review will be useful to broad audience of students, academicians and industry professionals who consider critical issues of protein delivery to the brain and aim developing and studying effective brain delivery systems for protein therapeutics.


Subject(s)
Blood-Brain Barrier/physiology , Central Nervous System Diseases/drug therapy , Drug Carriers , Peptides/therapeutic use , Proteins/therapeutic use , Blood-Brain Barrier/pathology , Humans , Nanoparticles
18.
Free Radic Biol Med ; 73: 299-307, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24924945

ABSTRACT

Excessive production of superoxide (O2(-)) in the central nervous system has been widely implicated in the pathogenesis of cardiovascular diseases, including chronic heart failure and hypertension. In an attempt to overcome the failed therapeutic impact of currently available antioxidants in cardiovascular disease, we developed a nanomedicine-based delivery system for the O2(-)-scavenging enzyme copper/zinc superoxide dismutase (CuZnSOD), in which CuZnSOD protein is electrostatically bound to a poly-l-lysine (PLL50)-polyethylene glycol (PEG) block copolymer to form a CuZnSOD nanozyme. Various formulations of CuZnSOD nanozyme are covalently stabilized by either reducible or nonreducible crosslinked bonds between the PLL50-PEG polymers. Herein, we tested the hypothesis that PLL50-PEG CuZnSOD nanozyme delivers active CuZnSOD protein to neurons and decreases blood pressure in a mouse model of angiotensin II (AngII)-dependent hypertension. As determined by electron paramagnetic resonance spectroscopy, nanozymes retain full SOD enzymatic activity compared to native CuZnSOD protein. Nonreducible CuZnSOD nanozyme delivers active CuZnSOD protein to central neurons in culture (CATH.a neurons) without inducing significant neuronal toxicity. Furthermore, in vivo studies conducted in adult male C57BL/6 mice demonstrate that hypertension established by chronic subcutaneous infusion of AngII is significantly attenuated for up to 7 days after a single intracerebroventricular injection of nonreducible nanozyme. These data indicate the efficacy of nonreducible PLL50-PEG CuZnSOD nanozyme in counteracting excessive O2(-) and decreasing blood pressure in AngII-dependent hypertensive mice after central administration. Additionally, this study supports the further development of PLL50-PEG CuZnSOD nanozyme as an antioxidant-based therapeutic option for hypertension.


Subject(s)
Drug Delivery Systems , Free Radical Scavengers/pharmacology , Hypertension/drug therapy , Superoxide Dismutase/administration & dosage , Superoxide Dismutase/pharmacology , Angiotensin II/metabolism , Animals , Antioxidants/pharmacology , Cells, Cultured , Central Nervous System , Electron Spin Resonance Spectroscopy , Heart Failure/drug therapy , Male , Mice , Mice, Inbred C57BL , Nanoparticles , Polyethylene Glycols , Polymers , Superoxides/metabolism
19.
Nanomedicine (Lond) ; 9(9): 1403-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24237263

ABSTRACT

AIMS: Active targeted transport of the nanoformulated redox enzyme, catalase, in macrophages attenuates oxidative stress and as such increases survival of dopaminergic neurons in animal models of Parkinson's disease. Optimization of the drug formulation is crucial for the successful delivery in living cells. We demonstrated earlier that packaging of catalase into a polyion complex micelle ('nanozyme') with a synthetic polyelectrolyte block copolymer protected the enzyme against degradation in macrophages and improved therapeutic outcomes. We now report the manufacture of nanozymes with superior structure and therapeutic indices. METHODS: Synthesis, characterization and therapeutic efficacy of optimal cell-based nanoformulations are evaluated. RESULTS: A formulation design for drug carriers typically works to avoid entrapment in monocytes and macrophages focusing on small-sized nanoparticles with a polyethylene glycol corona (to provide a stealth effect). By contrast, the best nanozymes for delivery in macrophages reported in this study have a relatively large size (≈ 200 nm), which resulted in improved loading capacity and release from macrophages. Furthermore, the cross-linking of nanozymes with the excess of a nonbiodegradable linker ensured their low cytotoxicity, and efficient catalase protection in cell carriers. Finally, the 'alternatively activated' macrophage phenotype (M2) utilized in these studies did not promote further inflammation in the brain, resulting in a subtle but statistically significant effect on neuronal regeneration and repair in vivo. CONCLUSION: The optimized cross-linked nanozyme loaded into macrophages reduced neuroinflammatory responses and increased neuronal survival in mice. Importantly, the approach for nanoformulation design for cell-mediated delivery is different from the common requirements for injectable formulations.


Subject(s)
Blood-Brain Barrier/metabolism , Catalase/administration & dosage , Drug Delivery Systems , Macrophages/physiology , Nanocapsules/administration & dosage , Animals , Catalase/metabolism , Cattle , Cell Line , Chemistry, Pharmaceutical , Cross-Linking Reagents , Drug Carriers/chemistry , Encephalitis/drug therapy , Encephalitis/pathology , Encephalitis/physiopathology , Enzyme Stability , Macrophage Activation , Macrophages/cytology , Macrophages/enzymology , Male , Mice , Mice, Inbred C57BL , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Nanomedicine , Particle Size
20.
Am J Physiol Regul Integr Comp Physiol ; 304(11): R917-28, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23552499

ABSTRACT

Cardiovascular diseases, including hypertension and heart failure, are associated with activation of the renin-angiotensin system (RAS) and increased circulating and tissue levels of ANG II, a primary effector peptide of the RAS. Through its actions on various cell types and organ systems, ANG II contributes to the pathogenesis of cardiovascular diseases by inducing cardiac and vascular hypertrophy, vasoconstriction, sodium and water reabsorption in kidneys, sympathoexcitation, and activation of the immune system. Cardiovascular research over the past 15-20 years has clearly implicated an important role for elevated levels of reactive oxygen species (ROS) in mediating these pathophysiological actions of ANG II. As such, the use of antioxidants, to reduce the elevated levels of ROS, as potential therapies for various ANG II-associated cardiovascular diseases has been intensely investigated. Although some antioxidant-based therapies have shown therapeutic impact in animal models of cardiovascular disease and in human patients, others have failed. In this review, we discuss the benefits and limitations of recent strategies, including gene therapy, dietary sources, low-molecular-weight free radical scavengers, polyethylene glycol conjugation, and nanomedicine-based technologies, which are designed to deliver antioxidants for the improved treatment of cardiovascular diseases. Although much work has been completed, additional research focusing on developing specific antioxidant molecules or proteins and identifying the ideal in vivo delivery system for such antioxidants is necessary before the use of antioxidant-based therapies for cardiovascular diseases become a clinical reality.


Subject(s)
Angiotensin II/physiology , Antioxidants/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Diet , Genetic Therapy , Humans , Nanoparticles , Reactive Oxygen Species , Renin-Angiotensin System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...