Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
ACS Nano ; 9(3): 2876-85, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25712076

ABSTRACT

Engineered particles adsorb biomolecules (e.g., proteins) when introduced in a biological medium to form a layer called a "corona". Coronas, in particular the protein corona, play an important role in determining the surface properties of particles and their targeting abilities. This study examines the influence of protein coronas on the targeting ability of layer-by-layer (LbL)-assembled polymer capsules and core-shell particles functionalized with monoclonal antibodies. Upon exposure of humanized A33 monoclonal antibody (huA33 mAb)-functionalized poly(methacrylic acid) (PMA) capsules or huA33 mAb-PMA particles to human serum, a total of 83 or 65 proteins were identified in the protein coronas, respectively. Human serum of varying concentrations altered the composition of the protein corona. The antibody-driven specific cell membrane binding was qualitatively and quantitatively assessed by flow cytometry and fluorescence microscopy in both the absence and presence of a protein corona. The findings show that although different protein coronas formed in human serum (at different concentrations), the targeting ability of both the huA33 mAb-functionalized PMA capsules and particles toward human colon cancer cells was retained, demonstrating no significant difference compared with capsules and particles in the absence of protein coronas: ∼70% and ∼90% A33-expressing cells were targeted by the huA33 mAb-PMA capsules and particles, respectively, in a mixed cell population. This result demonstrates that the formation of protein coronas did not significantly influence the targeting ability of antibody-functionalized LbL-polymer carriers, indicating that the surface functionality of engineered particles in the presence of protein coronas can be preserved.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Drug Carriers/chemistry , Protein Corona/chemistry , Adsorption , Antibodies, Monoclonal, Humanized/immunology , Blood Proteins/chemistry , Cell Line, Tumor , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Models, Molecular , Polymethacrylic Acids/chemistry , Protein Conformation
2.
Biomacromolecules ; 15(1): 53-9, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24328262

ABSTRACT

Thermodynamically assembled core-shell nanocarriers are potential candidates for drug delivery applications due to their submicrometer size and the ability to load drugs into their hydrophobic core. Herein, we describe the formation of core-shell particles that consist of noncovalent polymers, that is, polyrotaxanes (PRXs), that form an α-cyclodextrin (αCD) core surrounded by a corona of low-fouling poly(ethylene glycol) (PEG). The PRX core-shell particles are able to sequester small organic molecules, such as pyrene and calcein, releasing these small molecules during degradation. The small, cellular peptide, glutathione, was used to degrade the particles through the reductive cleavage of disulfide bonds that stabilize the individual PRX polymers. Cleavage of a single bond allows for the degradation of the supramolecular-polymer, making these PRX core-shell particles highly responsive. Furthermore, these particles demonstrate negligible cytotoxicity in mammalian cells, making them promising carriers for future drug delivery research.


Subject(s)
Cyclodextrins/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Poloxamer/chemistry , Rotaxanes/chemistry , Cell Survival/drug effects , Cell Survival/physiology , Cyclodextrins/pharmacology , Dose-Response Relationship, Drug , Drug Carriers/pharmacology , HeLa Cells , Humans , Poloxamer/pharmacology , Rotaxanes/pharmacology
3.
ACS Nano ; 7(12): 10960-70, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24256422

ABSTRACT

Many biomolecules, mainly proteins, adsorb onto polymer particles to form a dynamic protein corona in biological environments. The protein corona can significantly influence particle-cell interactions, including internalization and pathway activation. In this work, we demonstrate the differential roles of a given protein corona formed in cell culture media in particle uptake by monocytes and macrophages. By exposing disulfide-stabilized poly(methacrylic acid) nanoporous polymer particles (PMASH NPPs) to complete cell growth media containing 10% fetal bovine serum, a protein corona, with the most abundant component being bovine serum albumin, was characterized. Upon adsorption onto the PMASH NPPs, native bovine serum albumin (BSA) was found to undergo conformational changes. The denatured BSA led to a significant decrease in internalization efficiency in human monocytic cells, THP-1, compared with the bare particles, due to reduced cell membrane adhesion. In contrast, the unfolded BSA on the NPPs triggered class A scavenger receptor-mediated phagocytosis in differentiated macrophage-like cells (dTHP-1) without a significant impact on the overall internalization efficiency. Taken together, this work demonstrates the disparate effects of a given protein corona on particle-cell interactions, highlighting the correlation between protein corona conformation in situ and relevant biological characteristics for biological functionalities.


Subject(s)
Macrophages/drug effects , Monocytes/drug effects , Nanopores , Polymers/chemistry , Proteins/chemistry , Adsorption , Animals , Cattle , Cell Line , Cell Membrane/metabolism , Disulfides/chemistry , Humans , Inflammation , Kinetics , Monocytes/cytology , Nanotechnology , Particle Size , Phagocytosis , Polymethacrylic Acids/chemistry , Protein Conformation , Serum Albumin, Bovine/chemistry , U937 Cells
4.
ACS Nano ; 7(6): 5558-67, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23713907

ABSTRACT

Nanoporous polymer particles (NPPs) prepared by mesoporous silica templating show promise as a new class of versatile drug/gene delivery vehicles owning to their high payload capacity, functionality, and responsiveness. Understanding the cellular dynamics of such particles, including uptake, intracellular trafficking, and distribution, is an important requirement for their development as therapeutic carriers. Herein, we examine the spatiotemporal map of the cellular processing of submicrometer-sized disulfide-bonded poly(methacrylic acid) (PMASH) NPPs in HeLa cells using both flow cytometry and fluorescence microscopy. The data show that the PMASH NPPs are transported from the early endosomes to the lysosomes within a few minutes. Upon cell division, the lysosome-enclosed PMASH NPPs are distributed asymmetrically between two daughter cells. Statistical analysis of cells during cytokinesis suggests that partitioning of particles is biased with an average segregation deviation of 60%. Further, two-dimensional difference gel electrophoresis (2D-DIGE) analysis reveals that 127 out of 3059 identified spots are differentially regulated upon exposure to the PMASH NPPs. Pathway analysis of the proteomics data suggests that ubiquitylation, a reversible modification of cellular proteins with ubiquitin, plays a central role in overall cellular responses to the particles. These results provide important insights into the cellular dynamics and heterogeneity of NPPs, as well as the mechanisms that regulate the motility of these particles within cells, all of which have important implications for drug susceptibility characteristics in cancer cells using particle-based carriers.


Subject(s)
Intracellular Space/metabolism , Mitosis , Nanoparticles , Polymethacrylic Acids/chemistry , Polymethacrylic Acids/metabolism , Biological Transport , Fluorescent Dyes/metabolism , HeLa Cells , Humans
6.
ACS Nano ; 6(8): 6667-74, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22872125

ABSTRACT

The development of nanoengineered particles, such as polymersomes, liposomes, and polymer capsules, has the potential to offer significant advances in vaccine and cancer therapy. However, the effectiveness of these carriers has the potential to be greatly improved if they can be specifically delivered to target cells. We describe a general method for functionalizing nanoengineered polymer capsules with antibodies using click chemistry and investigate their interaction with cancer cells in vitro. The binding efficiency to cells was found to be dependent on both the capsule-to-cell ratio and the density of antibody on the capsule surface. In mixed cell populations, more than 90% of target cells bound capsules when the capsule-to-target cell ratio was 1:1. Strikingly, greater than 50% of target cells exhibited capsules on the cell surface even when the target cells were present as less than 0.1% of the total cell population. Imaging flow cytometry was used to quantify the internalization of the capsules, and the target cells were found to internalize capsules efficiently. However, the role of the antibody in this process was determined to enhance accumulation of capsules on the cell surface rather than promote endocytosis. This represents a significant finding, as this is the first study into the role antibodies play in internalization of such capsules. It also opens up the possibility of targeting these capsules to cancer cells using targeting molecules that do not trigger an endocytic pathway. We envisage that this approach will be generally applicable to the specific targeting of a variety of nanoengineered materials to cells.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Antigens, Neoplasm/immunology , Colonic Neoplasms/immunology , Membrane Glycoproteins/immunology , Nanocapsules/chemistry , Antibodies/administration & dosage , Cell Line, Tumor , Humans , Materials Testing , Nanocapsules/ultrastructure , Particle Size , Protein Binding
7.
Angew Chem Int Ed Engl ; 51(29): 7132-6, 2012 Jul 16.
Article in English | MEDLINE | ID: mdl-22744941

ABSTRACT

All sorted: The enzyme Sortase A was used to catalyze functionalization of PEGylated capsules with an activation-specific anti-platelet single-chain antibody (scFv). This enzymatic method allows fast, covalent, and site-directed functionalization of delivery vehicles under mild conditions. Activation-specific anti-platelet scFv-coated PEGylated capsules exhibited a high level of selective binding to thrombi, thus suggesting their potential for thrombosis therapy.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Click Chemistry , Cysteine Endopeptidases/metabolism , Drug Carriers/chemistry , Drug Delivery Systems , Polyethylene Glycols/chemistry , Single-Chain Antibodies/chemistry , Staphylococcus aureus/enzymology , Capsules/chemistry , Capsules/metabolism , Drug Carriers/metabolism , Humans , Polyethylene Glycols/metabolism , Single-Chain Antibodies/metabolism , Thrombosis/drug therapy
8.
Tissue Eng Part A ; 17(3-4): 381-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20807005

ABSTRACT

Porous, tubular, flexible, and elastic poly(trimethylene carbonate) (PTMC) scaffolds (length 8 cm and inner diameter 3 mm) for vascular tissue engineering were prepared by means of a dip-coating and particulate leaching procedure. Using NaCl as porogen, scaffolds with an average pore size of 110 µm and a porosity of 85% were obtained. Before leaching the salt, the structures were made creep-resistant by means of crosslinking at 25 kGy gamma irradiation. To increase the efficiency of cell seeding, the scaffolds were provided with a microporous outer layer of 0.2 mm with an average pore size of 28 µm and a porosity of 65% (total wall thickness 1 mm). Human smooth muscle cells (SMCs) were seeded in these scaffolds with an efficiency of 43%, as determined after 24 h cell adhesion. SMCs were cultured in the scaffolds up to 14 days under stationary conditions or under pulsatile flow conditions in a bioreactor (pressure 70-130 mmHg, 69 pulsations/min, and average wall shear rate 320 s(-1)). Although SMCs proliferated under both conditions, cell numbers were three to five times higher in case of dynamic culturing. This was qualitatively confirmed by means of histology. Also, in terms of mechanical properties, the dynamically cultured constructs performed better than the statically cultured constructs. After culturing for 14 days, the maximum tensile strengths of the constructs, determined in the radial direction, had increased from 0.16 MPa (unseeded scaffold) to 0.48 MPa (dynamic culturing) and 0.38 MPa (static culturing). The results of this study indicate that a potentially useful medial layer for tissue-engineered vascular grafts can be prepared by dynamic culturing of human SMCs seeded in porous tubular poly(trimethylene carbonate) scaffolds.


Subject(s)
Blood Vessels/growth & development , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Polyesters/chemical synthesis , Tissue Engineering/instrumentation , Tissue Scaffolds , Biocompatible Materials/chemical synthesis , Cell Culture Techniques/instrumentation , Cell Proliferation , Equipment Failure Analysis , Humans , Prosthesis Design
9.
J Am Chem Soc ; 132(45): 15881-3, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-20977221

ABSTRACT

Targeted delivery of drugs to specific cells allows a high therapeutic dose to be delivered to the target site with minimal harmful side effects. Combining targeting molecules with nanoengineered drug carriers, such as polymer capsules, micelles and polymersomes, has significant potential to improve the therapeutic delivery and index of a range of drugs. We present a general approach for functionalization of low-fouling, nanoengineered polymer capsules with antibodies using click chemistry. We demonstrate that antibody (Ab)-functionalized capsules specifically bind to colorectal cancer cells even when the target cells constitute less than 0.1% of the total cell population. This precise targeting offers promise for drug delivery applications.


Subject(s)
Antibodies/chemistry , Click Chemistry , Nanocapsules/chemistry , Neoplasms/drug therapy , Polymers/chemistry , Antibodies/administration & dosage , Azides/chemistry , Cell Line, Tumor , Flow Cytometry , Humans , Microscopy, Fluorescence , Nanocapsules/administration & dosage , Neoplasms/immunology , Polymers/administration & dosage , Polymers/chemical synthesis
10.
ACS Nano ; 4(5): 2928-36, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20420377

ABSTRACT

Understanding the interactions between drug carriers and cells is of importance to enhance the delivery of therapeutics. The release of therapeutics into different intracellular environments, such as the lysosomes or the cell cytoplasm, will impact their pharmacological activity. Herein, we investigate the intracellular fate of layer-by-layer (LbL)-assembled, submicrometer-sized polymer hydrogel capsules in a human colon cancer derived cell line, LIM1899. The cellular uptake of the disulfide-stabilized poly(methacrylic acid) (PMA(SH)) capsules by colon cancer cells is a time-dependent process. Confocal laser scanning microscopy and transmission electron microscopy reveal that the internalized capsules are deformed in membrane-enclosed compartments, which further mature to late endosomes or lysosomes. We further demonstrate the utility of these redox-responsive PMA(SH) capsules for the delivery of doxorubicin (DOX) to colon cancer cells. The DOX-loaded PMA(SH) capsules demonstrate a 5000-fold enhanced cytotoxicity in cell viability studies compared to free DOX.


Subject(s)
Colorectal Neoplasms/pathology , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Carriers/metabolism , Hydrogels/chemistry , Intracellular Space/metabolism , Polymers/chemistry , Biological Transport , Cell Line, Tumor , Cell Nucleus/metabolism , Diffusion , Disulfides/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Lysosomes/metabolism , Particle Size , Silicon Dioxide/chemistry
11.
Clin Cancer Res ; 11(2 Pt 1): 429-39, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15701825

ABSTRACT

PURPOSE: Prenylation is essential for membrane localization and participation of proteins in various signaling pathways. This study examined the role of farnesylated and geranylgeranylated proteins in the regulation of myeloma cell proliferation. EXPERIMENTAL DESIGN: Antiproliferative and apoptotic effects of various modulators of farnesylated and geranylgeranylated proteins were investigated in myeloma cells. RESULTS: Depletion of geranylgeranylpyrophosphate inhibited myeloma cell proliferation through accumulation of cells in G(1) phase of the cell cycle and loss of cells in S phase. In contrast, depletion of farnesylpyrophosphate had no or only minor effects. Furthermore, inhibition of geranylgeranyl transferase I activity was more effective in reducing myeloma cell growth when compared with inhibition of farnesyl transferase activity. This indicates that protein geranylgeranylation is important for myeloma cell proliferation and cell cycle progression through G(1). Geranylgeranylated target proteins involved in the control of proliferation include GTPases, such as Rac-1, Cdc42, and RhoA. Inhibition of Rho, Rac, and Cdc42 GTPases by toxin B reduced proliferation, without affecting cell viability, whereas specific inhibition of Rho GTPases by C3 exoenzyme was without effect. This suggests a role for Rac and/or Cdc42 GTPases in myeloma cell growth. Rac-1 activity was found in all myeloma cell lines and was suppressed by the depletion of intracellular pools of geranylgeranylpyrophosphate, whereas interleukin-6 rapidly induced Rac-1 activation. Furthermore, dominant-negative Tat-Rac-1 reduced myeloma cell proliferation, whereas constitutively active Tat-Rac-1 enhanced proliferation. CONCLUSION: These results indicate that protein geranylgeranylation is essential for myeloma cell proliferation and suggest that Rac-1 is a regulator of myeloma cell growth.


Subject(s)
Cell Proliferation , Multiple Myeloma/pathology , Protein Prenylation/physiology , Protein Processing, Post-Translational/drug effects , Aged , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Farnesyltranstransferase , Female , Genes, Dominant , Humans , Interleukin-6/pharmacology , Male , Middle Aged , Polyisoprenyl Phosphates/metabolism , Sesquiterpenes , Tumor Cells, Cultured , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
12.
Clin Cancer Res ; 9(15): 5735-48, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14654559

ABSTRACT

PURPOSE: Prenylation is essential for membrane localization and participation of proteins in various signaling pathways. The following study was conducted to examine the importance of protein farnesylation and geranylgeranylation for the regulation of lymphoma cell survival and proliferation. EXPERIMENTAL DESIGN: Lymphoma cells were treated with the beta-hydroxy-beta-methylglutaryl-CoA reductase inhibitor lovastatin, which inhibits protein farnesylation and geranylgeranylation by the depletion of intracellular pools of farnesylpyrophosphate and geranylgeranylpyrophosphate. In addition, farnesyl transferase and geranylgeranyl transferase activities were specifically inhibited by FTI-277 and GGTI-298, respectively. RESULTS: Only inhibition of geranylgeranylation by lovastatin led to reduction of cell viability in lymphoma cell lines and purified tumor cells from lymphoma patients in a time- and dose-dependent way. Reduction in the number of viable cells was mediated by both induction of apoptosis and inhibition of proliferation. In addition, GGTI-298 was more effective in induction of apoptosis and inhibition of proliferation than FTI-277. Apoptosis induced by inhibition of protein geranylgeranylation was associated with a reduction of Mcl-1 protein levels, collapse of the mitochondrial transmembrane potential, and caspase-3 activation. Inhibition of proliferation resulted from the induction of G(1) arrest. Furthermore, lovastatin at low concentrations sensitized lymphoma cells to dexamethasone, including cells resistant to this drug. CONCLUSION: These results indicate that protein geranylgeranylation is critical for the regulation of lymphoma tumor cell survival and proliferation and that pharmacological agents such as lovastatin or geranylgeranyl transferase inhibitors, alone or in combination with other drugs, may be useful in the treatment of lymphoma.


Subject(s)
Cell Division/physiology , Cell Survival/physiology , Diterpenes/metabolism , Protein Processing, Post-Translational/drug effects , Adult , Aged , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Lovastatin/pharmacology , Male , Middle Aged , Simvastatin/pharmacology
13.
Blood ; 102(9): 3354-62, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12855556

ABSTRACT

HMG-CoA reductase is the rate-limiting enzyme of the mevalonate pathway leading to the formation of cholesterol and isoprenoids such as farnesylpyrophosphate (FPP) and geranylgeranylpyrophosphate (GGPP). The inhibition of HMG-CoA reductase by lovastatin induced apoptosis in plasma cell lines and tumor cells from patients with multiple myeloma. Here we show that cotreatment with mevalonate or geranylgeranyl moieties, but not farnesyl groups, rescued myeloma cells from lovastatin-induced apoptosis. In addition, the inhibition of geranylgeranylation by specific inhibition of geranylgeranyl transferase I (GGTase I) induced the apoptosis of myeloma cells. Apoptosis triggered by the inhibition of geranylgeranylation was associated with reduction of Mcl-1 protein expression, collapse of the mitochondrial transmembrane potential, expression of the mitochondrial membrane protein 7A6, cytochrome c release from mitochondria into the cytosol, and stimulation of caspase-3 activity. These results imply that protein geranylgeranylation is critical for regulating myeloma tumor cell survival, possibly through regulating Mcl-1 expression. Our results show that pharmacologic agents such as lovastatin or GGTase inhibitors may be useful in the treatment of multiple myeloma.


Subject(s)
Apoptosis/drug effects , Multiple Myeloma/pathology , Neoplasm Proteins/biosynthesis , Protein Prenylation/drug effects , Proto-Oncogene Proteins c-bcl-2 , Alkyl and Aryl Transferases/antagonists & inhibitors , Bone Marrow Cells/pathology , Cell Line, Tumor , Diterpenes/pharmacology , Down-Regulation , Drug Antagonism , Enzyme Inhibitors/pharmacology , Humans , Lovastatin/pharmacology , Mitochondria/drug effects , Mitochondrial Proteins/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Protein Prenylation/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...