Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Nano ; 16(7): 10292-10301, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35653306

ABSTRACT

Bioorthogonal catalysis (BC) generates chemical reactions not present in normal physiology for the purpose of disease treatment. Because BC catalytically produces the desired therapy only at the site of disease, it holds the promise of site-specific treatment with little or no systemic exposure or side effects. Transition metals are typically used as catalytic centers in BC; however, solubility and substrate specificity typically necessitate a coordinating enzyme and/or stabilizing superstructure for in vivo application. We report the use of self-assembling, porous exoshells (tESs) to encapsulate and deliver an iron-containing reaction center for the treatment of breast cancer. The catalytic center is paired with indole-3-acetic acid (IAA), a natural product found in edible plants, which undergoes oxidative decarboxylation, via reduction of iron(III) to iron(II), to produce free radicals and bioactive metabolites. The tES encapsulation is critical for endocytic uptake of BC reaction centers and, when followed by administration of IAA, results in apoptosis of MDA-MB-231 triple negative cancer cells and complete regression of in vivo orthotopic xenograft tumors (p < 0.001, n = 8 per group). When Renilla luciferase (rLuc) is substituted for horseradish peroxidase (HRP), whole animal luminometry can be used to monitor in vivo activity.


Subject(s)
Biological Products , Nanoparticles , Neoplasms , Animals , Humans , Ferric Compounds , Horseradish Peroxidase/metabolism , Catalysis , Iron
2.
Nanomedicine ; 18: 169-178, 2019 06.
Article in English | MEDLINE | ID: mdl-30853651

ABSTRACT

Mammalian cell membranes are often incompatible with chemical modifications typically used to increase circulation half-life. Using cellular nanoghosts as a model, we show that proline-alanine-serine (PAS) peptide sequences expressed on the membrane surface can extend the circulation time of a cell membrane derived nanotherapeutic. Membrane expression of a PAS 40 repeat sequence decreased protein binding and resulted in a 90% decrease in macrophage uptake when compared with non-PASylated controls (P ≤ 0.05). PASylation also extended circulation half-life (t1/2 = 37 h) compared with non-PASylated controls (t1/2 = 10.5 h) (P ≤ 0.005), resulting in ~7-fold higher in vivo serum concentrations at 24 h and 48 h (P ≤ 0.005). Genetically engineered membrane expression of PAS repeats may offer an alternative to PEGylation and provide extended circulation times for cellular membrane-derived nanotherapeutics.


Subject(s)
Cell Membrane/metabolism , Nanoparticles/therapeutic use , Protein Engineering , Adsorption , Animals , Blood Proteins/metabolism , Dynamic Light Scattering , HEK293 Cells , Humans , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats , Surface Properties , Tissue Distribution
3.
Nat Commun ; 8(1): 1442, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29129910

ABSTRACT

The expression and stabilization of recombinant proteins is fundamental to basic and applied biology. Here we have engineered a thermostable protein nanoparticle (tES) to improve both expression and stabilization of recombinant proteins using this technology. tES provides steric accommodation and charge complementation to green fluorescent protein (GFPuv), horseradish peroxidase (HRPc), and Renilla luciferase (rLuc), improving the yields of functional in vitro folding by ~100-fold. Encapsulated enzymes retain the ability to metabolize small-molecule substrates, presumably via four 4.5-nm pores present in the tES shell. GFPuv exhibits no spectral shifts in fluorescence compared to a nonencapsulated control. Thermolabile proteins internalized by tES are resistant to thermal, organic, chaotropic, and proteolytic denaturation and can be released from the tES assembly with mild pH titration followed by proteolysis.


Subject(s)
Archaeoglobus fulgidus/genetics , Escherichia coli/genetics , Green Fluorescent Proteins/metabolism , Horseradish Peroxidase/metabolism , Luciferases, Renilla/metabolism , Recombinant Proteins/biosynthesis , Archaeoglobus fulgidus/metabolism , Escherichia coli/metabolism , Gene Expression/physiology , Nanoparticles/metabolism , Protein Folding , Recombinant Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...