Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Adv Mater ; 32(43): e2004901, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32924219

ABSTRACT

The COVID-19 pandemic has taken a significant toll on people worldwide, and there are currently no specific antivirus drugs or vaccines. Herein it is a therapeutic based on catalase, an antioxidant enzyme that can effectively breakdown hydrogen peroxide and minimize the downstream reactive oxygen species, which are excessively produced resulting from the infection and inflammatory process, is reported. Catalase assists to regulate production of cytokines, protect oxidative injury, and repress replication of SARS-CoV-2, as demonstrated in human leukocytes and alveolar epithelial cells, and rhesus macaques, without noticeable toxicity. Such a therapeutic can be readily manufactured at low cost as a potential treatment for COVID-19.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Betacoronavirus/drug effects , Catalase/therapeutic use , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Animals , Anti-Inflammatory Agents/pharmacokinetics , Antioxidants/pharmacokinetics , Betacoronavirus/physiology , COVID-19 , Catalase/pharmacokinetics , Cell Line , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/virology , Macaca mulatta , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects , Pandemics , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/virology , SARS-CoV-2 , Virus Replication/drug effects
2.
Int J Pharm ; 582: 119321, 2020 May 30.
Article in English | MEDLINE | ID: mdl-32289483

ABSTRACT

Combining functional proteins with small molecular drugs into one entity may endow distinct synergistic advantages. However, on account of completely different physicochemical properties of such payloads, co-delivery through systemic administration for therapeutic purpose is challenging. Herein, we designed the protein-drug conjugate HSAP-DC-CAT (human serum albumin/Pt (IV)-dibenzocyclooctyne/chlorin e6-catalase) by modification of CAT and cisplatin pro-drug loaded HSA with pH-sensitive azide linker 3-(azidomethyl)-4-methyl-2,5-furandione (AzMMMan) followed by click chemistry assembly with DC. The dynamic covalent bonds between linker and proteins, on the one hand, can bridge proteins and small molecular drugs in the intermediate state for systemic delivery in the harsh in vivo environment; on the other hand, it can trigger traceless cleavage and release of drugs and proteins with full bioactivity in acidic microenvironment of tumor. The multifunctional HSAP-DC-CAT provides efficient cytosolic transduction in vitro, excellent blood half-lives after systemic administration, and significant antitumor outcome via integrated cisplatin-based chemotherapy and Ce6-based photodynamic therapy enhanced by catalase-induced manipulation of tumor hypoxia microenvironment. This study describes a universal formulation strategy for protein and small molecular drug by a bifunctional linker through amide reaction and click chemistry, with traceless in vivo release of therapeutic units.


Subject(s)
Antioxidants/pharmacology , Breast Neoplasms/drug therapy , Catalase/pharmacology , Cisplatin/pharmacology , Photochemotherapy , Porphyrins/pharmacology , Prodrugs/pharmacology , Radiation-Sensitizing Agents/pharmacology , Serum Albumin, Human/chemistry , Animals , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catalase/chemistry , Catalase/pharmacokinetics , Cell Line, Tumor , Chlorophyllides , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Click Chemistry , Delayed-Action Preparations , Drug Carriers , Drug Compounding , Female , Hydrogen-Ion Concentration , Mice, Nude , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacokinetics , Tumor Hypoxia , Tumor Microenvironment
3.
Small ; 15(46): e1903628, 2019 11.
Article in English | MEDLINE | ID: mdl-31577387

ABSTRACT

It is hard for current radionuclide therapy to render solid tumors desirable therapeutic efficacy owing to insufficient tumor-targeted delivery of radionuclides and severe tumor hypoxia. In this study, a biocompatible hybrid protein nanoreactor composed of human serum albumin (HSA) and catalase (CAT) molecules is constructed via glutaraldehyde-mediated crosslinking. The obtained HSA-CAT nanoreactors (NRs) show retained and well-protected enzyme stability in catalyzing the decomposition of H2 O2 and enable efficient labeling of therapeutic radionuclide iodine-131 (131 I). Then, it is uncovered that such HSA-CAT NRs after being intravenously injected into tumor-bearing mice exhibit efficient passive tumor accumulation as vividly visualized under the fluorescence imaging system and gamma camera. As the result, such HSA-CAT NRs upon tumor accumulation would significantly attenuate tumor hypoxia by decomposing endogenous H2 O2 produced by cancer cells to molecular oxygen, and thereby remarkably improve the therapeutic efficacy of radionuclide 131 I. This study highlights the concise preparation of biocompatible protein nanoreactors with efficient tumor homing and hypoxia attenuation capacities, thus enabling greatly improved tumor radionuclide therapy with promising potential for future clinical translation.


Subject(s)
Catalase/metabolism , Iodine Radioisotopes/administration & dosage , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Oxygen/metabolism , Radiopharmaceuticals/administration & dosage , Serum Albumin, Human/metabolism , Animals , Catalase/pharmacokinetics , Cell Line, Tumor , Female , Humans , Hydrodynamics , Iodine Radioisotopes/therapeutic use , Mice, Nude , Nanoparticles/ultrastructure , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Serum Albumin, Human/pharmacokinetics , Tumor Hypoxia
4.
Biomaterials ; 140: 79-87, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28633046

ABSTRACT

Most potent therapeutics are unable to cross the blood-brain barrier following systemic administration, which necessitates the development of unconventional, clinically applicable drug delivery systems. With the given challenges, biologically active vehicles are crucial to accomplishing this task. We now report a new method for drug delivery that utilizes living cells as vehicles for drug carriage across the blood brain barrier. Cellular backpacks, 7-10 µm diameter polymer patches of a few hundred nanometers in thickness, are a potentially interesting approach, because they can act as drug depots that travel with the cell-carrier, without being phagocytized. Backpacks loaded with a potent antioxidant, catalase, were attached to autologous macrophages and systemically administered into mice with brain inflammation. Using inflammatory response cells enabled targeted drug transport to the inflamed brain. Furthermore, catalase-loaded backpacks demonstrated potent therapeutic effects deactivating free radicals released by activated microglia in vitro. This approach for drug carriage and release can accelerate the development of new drug formulations for all the neurodegenerative disorders.


Subject(s)
Antioxidants/administration & dosage , Brain/drug effects , Catalase/administration & dosage , Drug Carriers/metabolism , Drug Delivery Systems/methods , Inflammation/drug therapy , Macrophages/metabolism , Animals , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Caco-2 Cells , Catalase/pharmacokinetics , Catalase/therapeutic use , Cattle , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells
5.
Artif Cells Nanomed Biotechnol ; 45(5): 981-989, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27322626

ABSTRACT

Inflammatory bowel disease (IBD) is an inflammatory disorder of the digestive tract reported to be primarily caused by oxidative stress. In this study, alginate encapsulated nanoceramic carriers were designed to deliver acid labile antioxidant enzyme catalase orally. Complete system was characterized for size, loading efficiency, in vitro antioxidant assay and in vitro release. The prepared nanoceramic system was found to be spherical with diameter of 925 ± 6.81 nm. The in vitro release data followed the Higuchi model in acidic buffer whereas in alkaline pH sustained and almost first order release of enzyme was observed up to 6 h.


Subject(s)
Catalase/chemistry , Catalase/therapeutic use , Ceramics/chemistry , Drug Carriers/chemistry , Inflammatory Bowel Diseases/drug therapy , Nanostructures/chemistry , Biocatalysis , Biological Availability , Catalase/metabolism , Catalase/pharmacokinetics , Drug Liberation , Enzyme Stability , Hydrogen-Ion Concentration , Protein Conformation
6.
Nefrología (Madr.) ; 34(4): 483-490, jul.-ago. 2014.
Article in Spanish | IBECS | ID: ibc-129629

ABSTRACT

Background and objectives: Nephrotic syndrome is a chronic disease especially common in the childhood and adolescence. Reactive oxygen species (ROS) and free radicals have significant role in the pathogenesis of nephrotic syndrome. The aim of this study was to evaluate the effect of soy protein and genistein (main isoflavone of soybean) on renal antioxidant status of nephrotic rats. Methods: This study was done for 8 weeks on 40 adult male Sprague-Dawley rats divided into four groups of 10 rats each. Study groups included: 1-Control, 2-Nephrotic syndrome, 3-Nephrotic syndrome+soy protein diet and 4-Nephrotic syndrome+soy protein diet+genistein. Urine protein and urine creatinine were measured. After homogenization of kidney, total antioxidant capacities (TAC), activities of catalase enzyme, the concentration of malondialdehydes (MDA) and carbolynated proteins were determined spectrophotometrically. Pathological examination was done on kidneys with light microscope. Cell viability was evaluated with MTT assay on WEHI-164 fibro sarcoma cell line. The MMP2 enzyme activity was evaluated in different concentrations of genistein. Results: Total antioxidant capacity was significantly increased in soy genistein. Catalase activity was significantly increased in soy and soy genistein groups. Protein carbonyl and MDA were significantly lower in soy and soy genistein groups. The scores of pathological examination showed significant improvement in soy and soy genistein groups. Genistein decreased the proliferation of the WEHI-164 fibrosarcoma cell line. Conclusion: It seems that soy protein decreases kidney damages in nephrotic syndrome. Adding genistein to soy protein causes improvements in antioxidant status of kidney tissue. Genistein decreases proliferation of cell


Antecedentes y objetivos: El síndrome nefrótico es una enfermedad crónica especialmente común en la infancia y la adolescencia. Las especies reactivas del oxígeno (ERO) y los radicales libres desempeñan un papel importante en la patogénesis del síndrome nefrótico. El objetivo de este estudio es evaluar los efectos de la genisteína (principal isoflavona de la soja) y la proteína de soja en el estado antioxidante renal de ratas nefróticas. Métodos: Este estudio se llevó a cabo durante 8 semanas con 40 ratas Sprague-Dawley machos adultas, que fueron divididas en cuatro grupos de 10. Cada uno de los grupos de estudio incluía: 1 control, 2 con síndrome nefrótico, 3 con síndrome nefrótico más una dieta a base de proteína de soja y 4 con síndrome nefrótico más una dieta a base de proteína de soja más genisteína. Se midieron tanto los niveles de proteína como de creatinina en orina. Tras la homogenización del tejido renal, se calcularon mediante espectrofotometría la capacidad antioxidante total (CAT), la actividad de la enzima catalasa, la concentración de malondialdehidos (MDA) y las proteínas carboniladas. El examen patológico de los riñones se realizó con el microscopio óptico. Además, se evaluó la viabilidad celular con un ensayo de MTT de la línea celular de fibrosarcoma WEHI-164. También se evaluó la actividad de la enzima MMP2 con distintas concentraciones de genisteína. Resultados: La capacidad antioxidante total aumentó significativamente en las ratas que tenían una dieta de genisteína, al igual que la actividad de la catalasa en aquellas con una dieta de soja y genisteína. En cambio, los grupos carbonilo de las proteínas y los niveles de MDA fueron significativamente inferiores en los animales con una dieta de soja y de genisteína. El examen patológico reveló una mejora significativa en los grupos con dietas de soja y de genisteína. Asimismo, la genisteína disminuyó la proliferación de la línea celular de fibrosarcoma WEHI-164. Conclusión: Parece ser que la proteína de soja reduce los daños renales causados por el síndrome nefrótico. La adición de genisteína a la proteína de soja produce mejoras en el estado antioxidante del tejido renal. La genisteína disminuye la proliferación celular


Subject(s)
Animals , Rats , Soybean Proteins/pharmacokinetics , Genistein/pharmacokinetics , Nephrotic Syndrome/drug therapy , Disease Models, Animal , Protective Agents/pharmacokinetics , Antioxidants/pharmacokinetics , Catalase/pharmacokinetics
7.
J. physiol. biochem ; 68(1): 37-45, mar. 2012.
Article in English | IBECS | ID: ibc-122376

ABSTRACT

No disponible


Legumes are considered to have beneficial health implications, which have been attributed to their phytochemical content. Polyphenols are considered the most important phytochemical compounds extensively studied for their antioxidant properties. The aim of the present study was to examine the effects of potent antioxidant legume plant extracts on xanthine oxidase (XO), catalase (CAT) and superoxide dismutase (SOD) activities. XO exerts a dual role, as it is the major contributor of free radicals during exercise while it generates uric acid, the most potent antioxidant molecule in plasma. CAT and SOD are two of the main enzymes of the antioxidant defence of tissues. We demonstrate that the majority of the extracts inhibited XO activity, but they had no effect on CAT inhibition and SOD induction when used at low concentrations. These results imply that the tested extracts may be considered as possible source of novel XO inhibitors. However, we have shown that allopurinol administration, a known XO inhibitor, before exercise reduces performance and induces oxidative stress in rats. Considering the fact that the extracts examined had an inhibitory effect on XO activity, possibly posing a restriction in their characterization as antioxidants, phytochemical antioxidant administration before exercise should probably be reconsidered (AU)


Subject(s)
Humans , Plant Extracts/pharmacokinetics , Xanthine Oxidase/pharmacokinetics , Polyphenols/pharmacokinetics , Catalase/pharmacokinetics , Superoxide Dismutase/pharmacokinetics , Protective Agents/pharmacokinetics , Antioxidant Response Elements , Fabaceae , Exercise/physiology
8.
Nanomedicine (Lond) ; 7(6): 815-33, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22236307

ABSTRACT

BACKGROUND: Macrophage-carried nanoformulated catalase ('nanozyme') attenuates neuroinflammation and protects nigrostriatal neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxication. This is facilitated by effective enzyme transfer from blood-borne macrophages to adjacent endothelial cells and neurons leading to the decomposition of reactive oxygen species. MATERIALS & METHODS: We examined the intra- and inter-cellular trafficking mechanisms of nanozymes by confocal microscopy. Improved neuronal survival mediated by nanozyme-loaded macrophages was demonstrated by fluorescence-activated cell sorting. RESULTS: In macrophages, nanozymes were internalized mainly by clathrin-mediated endocytosis then trafficked to recycling endosomes. The enzyme is subsequently released in exosomes facilitated by bridging conduits. Nanozyme transfer from macrophages to adjacent cells by endocytosis-independent mechanisms diffusing broadly throughout the recipient cells. In contrast, macrophage-free nanozymes were localized in lysosomes following endocytic entry. CONCLUSION: Facilitated transfer of nanozyme from cell to cell can improve neuroprotection against oxidative stress commonly seen during neurodegenerative disease processes.


Subject(s)
Brain/metabolism , Catalase/administration & dosage , Drug Delivery Systems/methods , Endosomes/metabolism , Macrophages/cytology , Nanoparticles/chemistry , Neurons/cytology , Animals , Brain/blood supply , Brain/cytology , Catalase/analysis , Catalase/pharmacokinetics , Cell Communication , Cell Line , Cells, Cultured , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Protein Transport
9.
J Pharm Sci ; 101(2): 552-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21953593

ABSTRACT

Removal of hydrogen peroxide by delivering catalase to the vicinity of metastasizing tumor cells is a promising approach for inhibiting tumor metastasis. To inhibit bone metastasis, catalase was conjugated with 3,5-di(ethylamino-2,2-bisphosphono)benzoic acid (Bip), a derivative of bone-seeking bisphosphonates, polyethylene glycol (PEG), or both. Bip-conjugated catalase derivatives, that is, catalase-Bip and PEG-catalase-Bip, exhibited a higher affinity for bone matrix as compared with their counterparts without Bip. The tissue distribution of (111) In-labeled catalase derivatives indicated that the accumulation of radioactivity in bones was increased by conjugation of either Bip or PEG with catalase. An experimental bone metastasis model was developed by injecting male C57BL/6 mice with murine melanoma B16-BL6/Luc cells, which stably express firefly luciferase into left ventricle. Repeated injections of catalase to tumor-bearing mice had no significant effect on the number of melanoma cells in tibiae and femurs, whereas injections of catalase-Bip, PEG-catalase, or PEG-catalase-Bip significantly reduced the number. These results indicate that targeted delivery of catalase to the bones can be achieved by conjugating the enzyme with either Bip or PEG, and this delivery is effective in inhibiting the bone metastasis of tumor cells.


Subject(s)
Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Catalase/pharmacology , Melanoma, Experimental/pathology , Animals , Catalase/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Tissue Distribution
10.
CNS Neurol Disord Drug Targets ; 10(8): 936-44, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22229327

ABSTRACT

Extremely low frequency electromagnetic fields (ELF-EMF) have been found to produce a variety of biological effects. These effects of ELF-EMF depend upon frequency, amplitude, and length of exposure, and are also related to intrinsic susceptibility and responsiveness of different cell types. Although the mechanism of this interaction is still obscure, ELF-EMF can influence cell proliferation, differentiation, cell cycle, apoptosis, DNA replication and protein expression. The aim of this study was to estimate various kinetic constants of catalase, cytochrome P450 and inducible nitric oxide synthase in response to ELF-EMF exposure in human HaCaT and THP-1 cell lines. In order to evaluate the effect of ELF-EMF on the modulation of cellular responses to an inflammatory stimulus, both cell lines were treated with lipopolysaccharide. To the best of our knowledge there is no available report on such type of kinetic study of selected enzymes in response to ELF-EMF in these cell lines. Therefore, the current study may reveal novel mechanism of ELFEMF biological interaction with the enzymological and hormonal systems of living organisms. These new insights may be important for ELF-EMF application particularly for wound healing, tissue regeneration, Parkinson's and Alzheimer's diseases.


Subject(s)
Catalase/pharmacokinetics , Cytochrome P-450 Enzyme System/pharmacokinetics , Electromagnetic Fields , Nitric Oxide Synthase Type II/pharmacokinetics , Cell Line, Transformed , Cell Line, Tumor , Enzyme Activation/physiology , Humans , Kinetics
11.
Nanomedicine (Lond) ; 5(3): 379-96, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20394532

ABSTRACT

BACKGROUND: Parkinson's disease is a common progressive neurodegenerative disorder associated with profound nigrostriatal degeneration. Regrettably, no therapies are currently available that can attenuate disease progression. To this end, we developed a cell-based nanoformulation delivery system using the antioxidant enzyme catalase to attenuate neuroinflammatory processes linked to neuronal death. METHODS: Nanoformulated catalase was obtained by coupling catalase to a synthetic polyelectrolyte of opposite charge, leading to the formation of a polyion complex micelle. The nanozyme was loaded into bone marrow macrophages and its transport to the substantia nigra pars compacta was evaluated in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. RESULTS: Therapeutic efficacy of bone marrow macrophages loaded with nanozyme was confirmed by twofold reductions in microgliosis as measured by CD11b expression. A twofold increase in tyrosine hydroxylase-expressing dopaminergic neurons was detected in nanozyme-treated compared with untreated MPTP-intoxicated mice. Neuronal survival was confirmed by magnetic resonance spectroscopic imaging. Bone marrow macrophage-loaded catalase showed sustained release of the enzyme in plasma. CONCLUSION: These data support the importance of macrophage-based nanozyme carriage for Parkinson's disease therapies.


Subject(s)
Catalase/administration & dosage , Catalase/therapeutic use , Macrophages/drug effects , Nanostructures , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Brain/drug effects , Catalase/pharmacokinetics , Cell Adhesion/drug effects , Cell Movement/drug effects , Drug Carriers/chemistry , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanostructures/chemistry , Neuroprotective Agents/pharmacokinetics , Oxidative Stress/drug effects
12.
J Control Release ; 137(2): 110-5, 2009 Jul 20.
Article in English | MEDLINE | ID: mdl-19361547

ABSTRACT

Catalase delivery can be effective in inhibiting reactive oxygen species (ROS)-mediated acceleration of tumor metastasis. Our previous studies have demonstrated that increasing the plasma half-life of catalase by pegylation (PEG-catalase) significantly increases its potency of inhibiting experimental pulmonary metastasis in mice. In the present study, a biodegradable gelatin hydrogel formulation was used to further increase the circulation time of PEG-catalase. Implantation of (111)In-PEG-catalase/hydrogel into subcutaneous tissues maintained the radioactivity in plasma for more than 14 days. Then, the effect of the PEG-catalase/hydrogel on spontaneous pulmonary metastasis of tumor cells was evaluated in mice with subcutaneous tumor of B16-BL6/Luc cells, a murine melanoma cell line stably expressing luciferase. Measuring luciferase activity in the lung revealed that the PEG-catalase/hydrogel significantly (P<0.05) inhibited the pulmonary metastasis compared with PEG-catalase solution. These findings indicate that sustaining catalase activity in the blood circulation achieved by the use of pegylation and gelatin hydrogel can reduce the incidence of tumor cell metastasis.


Subject(s)
Antineoplastic Agents/administration & dosage , Catalase/administration & dosage , Catalase/therapeutic use , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/prevention & control , Melanoma, Experimental/secondary , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Catalase/chemistry , Catalase/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Gelatin/administration & dosage , Gelatin/chemistry , Gelatin/pharmacokinetics , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacokinetics , Injections, Subcutaneous , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/drug therapy , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics
13.
Pharm Res ; 26(1): 250-60, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18956141

ABSTRACT

PURPOSE: Based on a unique phase alignment that occurs during formulation, we postulated that PEG-ylation of the cargo enzyme would enhance its encapsulation within diblock copolymer nanocarriers and thus resistance to proteases. METHODS: A freeze-thaw modified double emulsion technique was utilized to encapsulate either the catalytically active enzyme catalase (MW approximately 250 kDa) or PEG-catalase in PEG-PLA polymer nanocarriers (PNC). Spectrophotometer measurement of substrate depletion was utilized to monitor enzyme activity. Isotope labeling of the enzyme was used in conjunction with activity measurements to determine PNC loading efficiency and PNC-enzyme resistance to proteases. This labeling also enabled blood clearance measurements of PNC-loaded and non-loaded enzymes in mice. RESULTS: Non-loaded PEG-catalase exhibited longer circulation times than catalase, but was equally susceptible to proteolysis. Modulation of the ratio of relatively hydrophilic to hydrophobic domains in the diblock PEG-PLA copolymer provided either filamentous or spherical PNC loaded with PEG-catalase. For both PNC geometries, encapsulation and resistance to proteases of the resultant PNC-loaded enzyme were more effective for PEG-catalase than catalase. Isotope tracing showed similar blood levels of PNC-loaded and free PEG-catalase in mice. CONCLUSIONS: PEGylation enhances active catalase loading within PNC and resistance to protease degradation, relative to unloaded PEG-catalase.


Subject(s)
Catalase/chemistry , Catalase/pharmacokinetics , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Animals , Chemistry, Pharmaceutical , Diffusion , Drug Carriers/chemistry , Freezing , Hydrogen Peroxide/chemistry , Indicators and Reagents , Lactic Acid/chemistry , Mice , Mice, Inbred C57BL , Microspheres , Nanotubes , Particle Size , Peptide Hydrolases/chemistry , Polyesters , Polymers/chemistry , Tissue Distribution
14.
Pharm Res ; 26(1): 244-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18548338

ABSTRACT

PURPOSE: To determine if particle shape can be engineered to inhibit phagocytosis of drug delivery particles by macrophages, which can be a significant barrier to successful therapeutic delivery. METHODS: Non-spherical polystyrene particles were fabricated by stretching spherical particles embedded in a polymer film. A rat alveolar macrophage cell line was used as model macrophages. Phagocytosis of particles was assessed using time-lapse video microscopy and fluorescence microscopy. RESULTS: We fabricated worm-like particles with very high aspect ratios (>20). This shape exhibits negligible phagocytosis compared to conventional spherical particles of equal volume. Reduced phagocytosis is a result of decreasing high curvature regions of the particle to two single points, the ends of the worm-like particles. Internalization is possible only at these points, while attachment anywhere along the length of the particles inhibits internalization due to the low curvature. CONCLUSIONS: Shape-induced inhibition of phagocytosis of drug delivery particles is possible by minimizing the size-normalized curvature of particles. We have created a high aspect ratio shape that exhibits negligible uptake by macrophages.


Subject(s)
Catalase/administration & dosage , Catalase/chemistry , Drug Carriers , Nanoparticles/chemistry , Nanotubes/chemistry , Polyethylene Glycols/chemistry , Animals , Catalase/pharmacokinetics , Chemistry, Pharmaceutical , Drug Compounding , Emulsions , Excipients , Freezing , Hydrogen Peroxide/chemistry , Indicators and Reagents , Mice , Microscopy, Electron, Scanning , Particle Size , Peptide Hydrolases/chemistry , Tissue Distribution
15.
Arch Pharm (Weinheim) ; 339(7): 372-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16838281

ABSTRACT

Bovine liver catalase (EC 1.11.1.6) was chemically modified with mannan, carboxymethylcellulose, and carboxymethylchitin. The enzyme retained about 48-97% of the initial specific activity after glycosidation with the polysaccharides. The prepared neoglycoenzyme was 1.9-5.7 fold more stable against the thermal inactivation processes at 55 degrees C, in comparison with the native counterpart. Also, the modified enzyme was more resistant to proteolytic degradation with trypsin. Pharmacokinetics studies revealed higher plasma half-life time for all the enzyme-polymer preparations, but better results were achieved for the enzyme modified with the anionic macromolecules.


Subject(s)
Catalase/pharmacokinetics , Polysaccharides/chemistry , Animals , Area Under Curve , Carbohydrate Sequence , Carboxymethylcellulose Sodium/chemistry , Carboxymethylcellulose Sodium/metabolism , Catalase/administration & dosage , Catalase/chemistry , Cattle , Enzyme Stability , Half-Life , Hot Temperature , Hydrogen-Ion Concentration , Injections, Intravenous , Liver/enzymology , Male , Molecular Sequence Data , Molecular Structure , Rats , Rats, Wistar , Solubility , Spectrophotometry , Substrate Specificity , Water/chemistry
16.
J Biotechnol ; 122(3): 326-33, 2006 Apr 10.
Article in English | MEDLINE | ID: mdl-16446004

ABSTRACT

An enzymatic approach, based on a transglutaminase-catalyzed coupling reaction, was investigated to modify bovine liver catalase with an end-group aminated dextran derivative. We demonstrated that catalase activity increased after enzymatic glycosidation and that the conjugate was 3.8-fold more stable to thermal inactivation at 55 degrees C and 2-fold more resistant to proteolytic degradation by trypsin. Moreover, the transglutaminase-mediated modification also improved the pharmacokinetics behavior of catalase, increasing 2.5-fold its plasma half-life time and reducing 3-fold the total clearance after its i.v. administration in rats.


Subject(s)
Catalase/chemistry , Dextrans/chemistry , Transglutaminases/chemistry , Animals , Cadaverine/analogs & derivatives , Cadaverine/chemistry , Catalase/blood , Catalase/pharmacokinetics , Catalysis , Cattle , Dextrans/pharmacokinetics , Diamines/chemistry , Female , Fluorescent Dyes/chemistry , Rats , Rats, Wistar , Streptomyces/enzymology
17.
J Control Release ; 109(1-3): 101-7, 2005 Dec 05.
Article in English | MEDLINE | ID: mdl-16256238

ABSTRACT

To develop effective anti-metastatic therapy, targeted or sustained delivery of catalase was examined in mice. We found that mouse lung with metastatic colonies of adenocarcinoma colon26 cells exhibited reduced catalase activity. The interaction of the tumor cells with macrophages or hepatocytes generated detectable amounts of ROS, and increased the activity of matrix metalloproteinases. Hepatocyte-targeted delivery of catalase was successfully achieved by galactosylation, which was highly effective in inhibiting the hepatic metastasis of colon26 cells. PEGylation, which increased the retention of catalase in the circulation, effectively inhibited the pulmonary metastasis of the cells. To examine which processes in tumor metastasis are inhibited by catalase derivatives, the tissue distribution and proliferation of tumor cells in mice was quantitatively analyzed using firefly luciferase-expressing tumor cells. An injection of PEG-catalase just before the inoculation of melanoma B16-BL6/Luc cells significantly reduced the number of the tumor cells in the lung at 24 h. Daily dosing of PEG-catalase greatly inhibited the proliferation of the tumor cells, and increased the survival rate of the tumor-bearing mice. These results indicate that targeted or sustained delivery of catalase to sites where tumor cells metastasize is a promising approach for inhibiting metastatic tumor growth.


Subject(s)
Antioxidants/therapeutic use , Drug Delivery Systems , Enzyme Therapy , Neoplasm Metastasis/therapy , Animals , Antioxidants/administration & dosage , Catalase/administration & dosage , Catalase/pharmacokinetics , Catalase/therapeutic use , Enzymes/administration & dosage , Humans , Mice , Tissue Distribution
18.
AAPS PharmSciTech ; 5(4): e51, 2004 Oct 11.
Article in English | MEDLINE | ID: mdl-15760048

ABSTRACT

The purpose of this research was to encapsulate superoxide dismutase (SOD) and catalase (CAT) in biodegradable microspheres (MS) to obtain suitable sustained protein delivery. A modified water/oil/water double emulsion method was used for poly(D,L-lactide-co-glycolide) (PLGA) and poly(D,L-lactide) PLA MS preparation co-encapsulating mannitol, trehalose, and PEG400 for protein stabilization. Size, morphology, porosity, mass loss, mass balance, in vitro release and in vitro activity were assessed by using BCA protein assay, scanning electron microscopy, BET surface area, and particle-sizing techniques. In vitro activity retention within MS was evaluated by nicotinammide adenine dinucleotide oxidation and H2O2 consumption assays. SOD encapsulation efficiency resulted in 30% to 34% for PLA MS and up to 51% for PLGA MS, whereas CAT encapsulation was 34% and 45% for PLGA and PLA MS, respectively. All MS were spherical with a smooth surface and low porosity. Particle mean diameters ranged from 10 to 17 mum. CAT release was prolonged, but the results were incomplete for both PLA and PLGA MS, whereas SOD was completely released from PLGA MS in a sustained manner after 2 months. CAT results were less stable and showed a stronger interaction than SOD with the polymers. Mass loss and mass balance correlated well with the release profiles. SOD and CAT in vitro activity was preserved in all the preparations, and SOD was better stabilized in PLGA MS. PLGA MS can be useful for SOD delivery in its native form and is promising as a new depot system.


Subject(s)
Catalase/pharmacokinetics , Drug Carriers/pharmacokinetics , Microspheres , Superoxide Dismutase/pharmacokinetics , Animals , Catalase/administration & dosage , Cattle , Drug Carriers/administration & dosage , Superoxide Dismutase/administration & dosage
19.
Int J Cancer ; 99(3): 474-9, 2002 May 20.
Article in English | MEDLINE | ID: mdl-11992420

ABSTRACT

In a previous study, we showed that targeted delivery of bovine liver catalase to hepatocytes by direct galactosylation augmented the inhibitory effect of the enzyme on experimental hepatic metastasis of colon carcinoma cells (unpublished data). Here, we examined the ability of catalase to inhibit tumor metastasis to the lung by controlling its biodistribution. Four types of catalase derivative, Gal-CAT, Man-CAT, Suc-CAT and PEG-CAT, were synthesized. Experimental pulmonary metastasis was induced in mice by i.v. injection of 1 x 10(5) colon 26 tumor cells. An i.v. injection of catalase (35,000 units/kg) partially, but significantly, decreased the number of colonies in the lung 2 weeks after tumor injection, from 93 +/- 29 (saline injection) to 63 +/- 23 (p < 0.01). Suc-CAT, Man-CAT and Gal-CAT showed effects similar to those of catalase on the number of colonies. However, PEG-CAT greatly inhibited pulmonary metastasis to 22 +/- 11 (p < 0.001). Furthermore, s.c. injection of catalase also greatly inhibited metastasis (11 +/- 6, p < 0.001). Neither inactivated catalase nor BSA showed any effects on the number of metastatic colonies, indicating that the enzymatic activity of catalase to detoxify H(2)O(2) is the critical factor inhibiting metastasis. (111)In-PEG-CAT showed a sustained concentration in plasma, whereas s.c.-injected (111)In-catalase was slowly absorbed from the injection site. These results suggest that retention of catalase activity in the circulation is a promising approach to inhibit pulmonary metastasis.


Subject(s)
Catalase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Animals , Catalase/pharmacokinetics , Cattle , Hepatocytes/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Indium/pharmacology , Liver/enzymology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Organ Size , Radioisotopes/pharmacology , Tissue Distribution , Tumor Cells, Cultured
20.
J Control Release ; 80(1-3): 169-78, 2002 Apr 23.
Article in English | MEDLINE | ID: mdl-11943396

ABSTRACT

A glucose-sensitive polymeric composite membrane was prepared based on our previously developed stimuli-responsive membrane system. Membranes were cast from a mixture of glucose oxidase (GOD), catalase, and poly(N-isopropylacrylamide-co-methacrylic acid) (poly(NIPAm/MAA)) nanoparticles dispersed in a solution of a hydrophobic polymer. High efficiency of enzyme immobilization was achieved with undetectable leakage. The bioactivity of the immobilized GOD, as measured by pH change of glucose solutions, was found to be equivalent to approximately 80% of that of the free GOD. The addition of catalase markedly increased the oxidation rate of glucose. However, an optimal unit ratio of GOD to catalase and optimal enzyme loading were observed. The rate of insulin permeation through the membrane was modulated by glucose concentration due to shrinking or swelling of the embedded pH-sensitive nanoparticles. The response of insulin permeability to the change in the glucose concentration could be detected within 5-15 min. The permeability of insulin increased more than 3-fold as the glucose concentration was raised from 50 to 200 mg/dl. The average insulin permeability at 400 mg/dl of glucose was 8-fold that at 50 mg/dl in a continuous test in saline and was 6-fold in a three-cycle discontinuous test in pH 7.4 buffer.


Subject(s)
Glucose/pharmacokinetics , Insulin/pharmacokinetics , Membranes, Artificial , Polymers/pharmacokinetics , Catalase/pharmacokinetics , Enzymes, Immobilized/pharmacokinetics , Glucose Oxidase/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Nanotechnology/methods , Permeability/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...