Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
BioTech (Basel) ; 12(1)2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36810440

ABSTRACT

Nanosized drug delivery systems (DDS) have been studied as a novel strategy against cancer due to their potential to simultaneously decrease drug inactivation and systemic toxicity and increase passive and/or active drug accumulation within the tumor(s). Triterpenes are plant-derived compounds with interesting therapeutic properties. Betulinic acid (BeA) is a pentacyclic triterpene that has great cytotoxic activity against different cancer types. Herein, we developed a nanosized protein-based DDS of bovine serum albumin (BSA) as the drug carrier combining two compounds, doxorubicin (Dox) and the triterpene BeA, using an oil-water-like micro-emulsion method. We used spectrophotometric assays to determine protein and drug concentrations in the DDS. The biophysical properties of these DDS were characterized using dynamic light scattering (DLS) and circular dichroism (CD) spectroscopy, confirming nanoparticle (NP) formation and drug loading into the protein structure, respectively. The encapsulation efficiency was 77% for Dox and 18% for BeA. More than 50% of both drugs were released within 24 h at pH 6.8, while less drug was released at pH 7.4 in this period. Co-incubation viability assays of Dox and BeA alone for 24 h demonstrated synergistic cytotoxic activity in the low µM range against non-small-cell lung carcinoma (NSCLC) A549 cells. Viability assays of the BSA-(Dox+BeA) DDS demonstrated a higher synergistic cytotoxic activity than the two drugs with no carrier. Moreover, confocal microscopy analysis confirmed the cellular internalization of the DDS and the accumulation of the Dox in the nucleus. We determined the mechanism of action of the BSA-(Dox+BeA) DDS, confirming S-phase cell cycle arrest, DNA damage, caspase cascade activation, and downregulation of epidermal growth factor receptor (EGFR) expression. This DDS has the potential to synergistically maximize the therapeutic effect of Dox and diminish chemoresistance induced by EGFR expression using a natural triterpene against NSCLC.

2.
BioTech (Basel) ; 11(2)2022 Mar 30.
Article in English | MEDLINE | ID: mdl-35822782

ABSTRACT

Botryococcus braunii (B. braunii) is a green microalga primarily found in freshwater, reservoirs, and ponds. Photosynthetic pigments from algae have shown many bioactive molecules with therapeutic potential. Herein, we report the purification, characterization, and anticancer properties of photosystem I light-harvesting complex I (PSI-LHCI) from the green microalga B. braunii UTEX2441. The pigment-protein complex was purified by sucrose density gradient and characterized by its distinctive peaks using absorption, low-temperature (77 K) fluorescence, and circular dichroism (CD) spectroscopic analyses. Protein complexes were resolved by blue native-PAGE and two-dimensional SDS-PAGE. Triple-negative breast cancer MDA-MB-231 cells were incubated with PSI-LHCI for all of our experiments. Cell viability was assessed, revealing a significant reduction in a time- and concentration-dependent manner. We confirmed the internalization of PSI-LHCI within the cytoplasm and nucleus after 12 h of incubation. Cell death mechanism by oxidative stress was confirmed by the production of reactive oxygen species (ROS) and specifically superoxide. Furthermore, we monitored autophagic flux, apoptotic and necrotic features after treatment with PSI-LHCI. Treated MDA-MB-231 cells showed positive autophagy signals in the cytoplasm and nucleus, and necrotic morphology by the permeabilization of the cell membrane. Our findings demonstrated for the first time the cytotoxic properties of B. braunii PSI-LHCI by the induction of ROS and autophagy in breast cancer cells.

3.
Pharmaceutics ; 14(3)2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35335867

ABSTRACT

The folate receptor alpha (FR), which is overexpressed in solid tumors including NSCLC, can be utilized for active tumor targeting to afford more effective cancer therapies. In this context, cytochrome c (Cyt c) has drawn attention to cancer research because it is non-toxic, yet, when delivered to the cytoplasm of cancer cells, can kill them by inducing apoptosis. Cyt c nanoparticles (NPs, 169 ± 9 nm) were obtained by solvent precipitation with acetonitrile, and stabilized by reversible homo-bifunctional crosslinking to accomplish a Cyt-c-based drug delivery system that combines stimulus-responsive release and active targeting. Cyt c was released under intracellular redox conditions, due to an S-S bond in the NPs linker, while NPs remained intact without any release under extracellular conditions. The NP surface was decorated with a hydrophilic folic acid-polyethylene glycol (FA-PEG) polymer for active targeting. The FA-decorated NPs specifically recognized and killed cancer cells (IC50 = 47.46 µg/mL) that overexpressed FR, but showed no toxicity against FR-negative cells. Confocal microscopy confirmed the preferential uptake and apoptosis induction of our NPs by FR-positive cancer cells. In vivo experiments using a Lewis lung carcinoma (LLC) mouse model showed visible NP accumulation within the tumor and inhibited the growth of LLC tumors.

4.
Cancer Drug Resist ; 4: 163-191, 2021.
Article in English | MEDLINE | ID: mdl-34142021

ABSTRACT

Cancer cells can develop resistance to anticancer drugs, thereby becoming tolerant to treatment through different mechanisms. The biological mechanisms leading to the generation of anticancer treatment resistance include alterations in transmembrane proteins, DNA damage and repair mechanisms, alterations in target molecules, and genetic responses, among others. The most common anti-cancer drugs reported to develop resistance to cancer cells include cisplatin, doxorubicin, paclitaxel, and fluorouracil. These anticancer drugs have different mechanisms of action, and specific cancer types can be affected by different genes. The development of drug resistance is a cellular response which uses differential gene expression, to enable adaptation and survival of the cell to diverse threatening environmental agents. In this review, we briefly look at the key regulatory genes, their expression, as well as the responses and regulation of cancer cells when exposed to anticancer drugs, along with the incorporation of alternative nanocarriers as treatments to overcome anticancer drug resistance.

5.
Inorganics (Basel) ; 9(11)2021 Nov.
Article in English | MEDLINE | ID: mdl-35978717

ABSTRACT

The heme protein cytochrome c (Cyt c) plays pivotal roles in cellular life and death processes. In the respiratory chain of mitochondria, it serves as an electron transfer protein, contributing to the proliferation of healthy cells. In the cell cytoplasm, it activates intrinsic apoptosis to terminate damaged cells. Insight into these mechanisms and the associated physicochemical properties and biomolecular interactions of Cyt c informs on the anticancer therapeutic potential of the protein, especially in its ability to subvert the current limitations of small molecule-based chemotherapy. In this review, we explore the development of Cyt c as an anticancer drug by identifying cancer types that would be receptive to the cytotoxicity of the protein and factors that can be finetuned to enhance its apoptotic potency. To this end, some information is obtained by characterizing known drugs that operate, in part, by triggering Cyt c induced apoptosis. The application of different smart drug delivery systems is surveyed to highlight important features for maintaining Cyt c stability and activity and improving its specificity for cancer cells and high drug payload release while recognizing the continuing limitations. This work serves to elucidate on the optimization of the strategies to translate Cyt c to the clinical market.

6.
ChemMedChem ; 15(22): 2185-2192, 2020 11 18.
Article in English | MEDLINE | ID: mdl-32918396

ABSTRACT

Malignant gliomas are the most lethal form of primary brain tumors. Despite advances in cancer therapy, the prognosis of glioma patients has remained poor. Cytochrome c (Cytc), an endogenous heme-based protein, holds tremendous potential to treat gliomas because of its innate capacity to trigger apoptosis. To this end, a hybrid cytochrome c-chlorotoxin (Cytc-CTX) protein was biosynthesized to enable cellular uptake of the cell impenetrable Cytc using CTX transporters. A nucleotide sequence containing 1 : 1 Cytc and CTX was constructed and separated by a hexahistidine-tag and an enterokinase cleavage site. The sequence was cloned into a pBTR1 plasmid, expressed in Escherichia coli, purified via 2-dimensional chromatography. The identity and size of the protein were determined by Western blot and mass spectrometry. Cytc in this soluble hybrid protein has similar structure and stability as human Cytc and the hybrid protein is endocytosed into a glioma cell line, while displaying potent cytotoxicity and a favorable therapeutic index. Its facile, low-cost, and high yield synthesis, biocompatibility, and robustness suggest that the hybrid protein is a promising candidate for antiglioma drug evaluation.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Cytochromes c/metabolism , Glioma/drug therapy , Scorpion Venoms/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Survival/drug effects , Cells, Cultured , Cytochromes c/chemistry , Dose-Response Relationship, Drug , Glioma/metabolism , Glioma/pathology , Humans , Scorpion Venoms/chemistry , Scorpion Venoms/metabolism , Structure-Activity Relationship
7.
Cancers (Basel) ; 12(5)2020 May 13.
Article in English | MEDLINE | ID: mdl-32413975

ABSTRACT

The delivery of Cytochrome c (Cyt c) to the cytosol stimulates apoptosis in cells where its release from mitochondria and apoptotic induction is inhibited. We developed a drug delivery system consisting of Cyt c nanoparticles decorated with folate-poly(ethylene glycol)-poly(lactic-co-glycolic acid)-thiol (FA-PEG-PLGA-SH) to deliver Cyt c into cancer cells and tested their targeting in the Lewis Lung Carcinoma (LLC) mouse model. Cyt c-PLGA-PEG-FA nanoparticles (NPs) of 253 ± 55 and 354 ± 11 nm were obtained by Cyt c nanoprecipitation, followed by surface decoration with the co-polymer SH-PLGA-PEG-FA. The internalization of Cyt c-PLGA-PEG-FA nanoparticles (NPs) in LLC cells was confirmed by confocal microscopy. NP caspase activation was more efficient than the NP-free formulation. Caspase activity assays showed NPs retained 88-96% Cyt c activity. The NP formulations were more effective in decreasing LLC cell viability than NP-free formulation, with IC50 49.2 to 70.1 µg/mL versus 129.5 µg/mL, respectively. Our NP system proved to be thrice as selective towards cancerous than normal cells. In vivo studies using near infrared-tagged nanoparticles show accumulation in mouse LLC tumor 5 min post-injection. In conclusion, our NP delivery system for Cyt c shows superiority over the NP-free formulation and reaches a folic acid-overexpressing tumor in an immune-competent animal model.

8.
Drug Des Devel Ther ; 13: 3753-3772, 2019.
Article in English | MEDLINE | ID: mdl-31802849

ABSTRACT

Cancer is the second largest cause of death worldwide with the number of new cancer cases predicted to grow significantly in the next decades. Biotechnology and medicine can and should work hand-in-hand to improve cancer diagnosis and treatment efficacy. However, success has been frequently limited, in particular when treating late-stage solid tumors. There still is the need to develop smart and synergistic therapeutic approaches to achieve the synthesis of strong and effective drugs and delivery systems. Much interest has been paid to the development of smart drug delivery systems (drug-loaded particles) that utilize passive targeting, active targeting, and/or stimulus responsiveness strategies. This review will summarize some main ideas about the effect of each strategy and how the combination of some or all of them has shown to be effective. After a brief introduction of current cancer therapies and their limitations, we describe the biological barriers that nanoparticles need to overcome, followed by presenting different types of drug delivery systems to improve drug accumulation in tumors. Then, we describe cancer cell membrane targets that increase cellular drug uptake through active targeting mechanisms. Stimulus-responsive targeting is also discussed by looking at the intra- and extracellular conditions for specific drug release. We include a significant amount of information summarized in tables and figures on nanoparticle-based therapeutics, PEGylated drugs, different ligands for the design of active-targeted systems, and targeting of different organs. We also discuss some still prevailing fundamental limitations of these approaches, eg, by occlusion of targeting ligands.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Humans
9.
Biomolecules ; 9(4)2019 04 18.
Article in English | MEDLINE | ID: mdl-31003476

ABSTRACT

In this study, we identified the proton-coupled folate transporter (PCFT) as a route for targeted delivery of drugs to some gliomas. Using the techniques of confocal imaging, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and small interfering (siRNA) knockdown against the PCFT, we demonstrated that Gl261 and A172 glioma cells, but not U87 and primary cultured astrocytes, express the PCFT, which provides selective internalization of folic acid (FA)-conjugated cytochrome c-containing nanoparticles (FA-Cyt c NPs), followed by cell death. The FA-Cyt c NPs (100 µg/mL), had no cytotoxic effects in astrocytes but caused death in glioma cells, according to their level of expression of PCFT. Whole-cell patch clamp recording revealed FA-induced membrane currents in FA-Cyt c NPs-sensitive gliomas, that were reduced by siRNA PCFT knockdown in a similar manner as by application of FA-Cyt c NPs, indicating that the PCFT is a route for internalization of FA-conjugated NPs in these glioma cells. Analysis of human glioblastoma specimens revealed that at least 25% of glioblastomas express elevated level of either PCFT or folate receptor (FOLR1). We conclude that the PCFT provides a mechanism for targeted delivery of drugs to some gliomas as a starting point for the development of efficient methods for treating gliomas with high expression of PCFT and/or FOLR1.


Subject(s)
Brain Neoplasms/metabolism , Cytochromes c/chemistry , Glioma/metabolism , Nanoconjugates/chemistry , Proton-Coupled Folate Transporter/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Cytochromes c/pharmacology , Folic Acid/chemistry , Folic Acid/pharmacology , Humans , Mice , Mice, Inbred C57BL , Nanoconjugates/adverse effects
10.
PLoS One ; 13(7): e0197643, 2018.
Article in English | MEDLINE | ID: mdl-30052638

ABSTRACT

L-Asparaginase is an enzyme successfully being used in the treatment of acute lymphoblastic leukemia, acute myeloid leukemia, and non-Hodgkin's lymphoma. However, some disadvantages still limit its full application potential, e.g., allergic reactions, pancreatitis, and blood clotting impairment. Therefore, much effort has been directed at improving its performance. A popular strategy is to randomly conjugate L-asparaginase with mono-methoxy polyethylene glycol, which became a commercial FDA approved formulation widely used in recent years. To improve this formulation by PEGylation, herein we performed cysteine-directed conjugation of the L-asparaginase subunits to prevent dissociation-induced loss of activity. The recombinant cysteine conjugation sites were introduced by mutagenesis at surface-exposed positions on the protein to avoid affecting the catalytic activity. Three conjugates were obtained using different linear PEGs of 1000, 2000, and 5000 g/mol, with physical properties ranging from a semi-solid gel to a fully soluble state. The soluble-conjugate exhibited higher catalytic activity than the non-conjugated mutant, and the same activity than the native enzyme. The cysteine-directed crosslinking of the L-asparaginase subunits produced a higher molecular weight conjugate compared to the native tetrameric enzyme. This strategy might improve L-asparaginase efficiency for leukemia treatment by reducing glomerular filtration due to the increase in hydrodynamic size thus extending half-live, while at the same time retaining full catalytic activity.


Subject(s)
Antineoplastic Agents/chemistry , Asparaginase/chemistry , Asparagine/chemistry , Cysteine/chemistry , Glycoconjugates/chemistry , Polyethylene Glycols/chemistry , Antineoplastic Agents/metabolism , Asparaginase/genetics , Asparaginase/metabolism , Asparagine/metabolism , Binding Sites , Biocatalysis , Cross-Linking Reagents/chemistry , Gene Expression , Glycoconjugates/genetics , Glycoconjugates/metabolism , Humans , Kinetics , Maleimides/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility , Substrate Specificity , Sulfhydryl Compounds/chemistry
11.
Curr Top Med Chem ; 18(5): 418-427, 2018.
Article in English | MEDLINE | ID: mdl-29766807

ABSTRACT

INTRODUCTION: The first total synthesis of ω-phenyl Δ6 fatty acids (FA) and their cytotoxicity (A549) and leishmanicidal (L. infantum) activities are described. The novel 16-phenyl-6-hexadecynoic acid (1) and the known 16-phenylhexadecanoic acid (2) were synthesized in 7-8 steps with overall yields of 46 % and 41 %, respectively. The syntheses of the unprecedented 10-phenyl-6-decynoic acid (3), 10-cyclohexyl-6-decynoic acid (4) and 10-(4-methoxyphenyl)-6-decynoic acid (5) was also performed in 3 steps with 73-76 % overall yields. The use of lithium acetylide coupling enabled the 4-step synthesis of 10-phenyl-6Z-decenoic acid (6) with a 100 % cis-stereochemistry. The cytotoxicity of these novel FA was determined against A549 cells and L. infantum promastigotes and amastigotes. Among the ω-phenylated FA, the best cytotoxicity towards A549 was displayed by 1, with an IC50 of 18 ± 1 µM. On the other hand, among the C10 acids, the ω-cyclohexyl acid 4 presented the best cytotoxicity (IC50 = 40 ± 2 µM) towards A549. RESULTS: Based on caspase-3/7 studies neither of the FA induced apoptosis in A549, thus implying other mechanisms of cell death. CONCLUSION: The antileishmanial studies were performed with the top Leishmania donovani topoisomerase IB (LdTopIB) inhibitors, namely 1 and 2 (EC50 between 14 and 36 µM, respectively), acids that did not stabilize the cleavage complexes between LdTopIB and DNA. Acids 1 and 2 displayed cytotoxicity towards L. infantum amastigotes (IC50 = 3-6 µM) and L. infantum promastigotes (IC50 = 60- 70 µM), but low toxicity towards murine splenocytes. Our results identified 1 as the optimum ω- phenylated acid of the series.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/pharmacology , Fatty Acids, Omega-6/chemical synthesis , Fatty Acids, Omega-6/pharmacology , Leishmania infantum/drug effects , Topoisomerase I Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Antiprotozoal Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Topoisomerases, Type I/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fatty Acids, Omega-6/chemistry , Humans , Molecular Structure , Parasitic Sensitivity Tests , Recombinant Proteins/metabolism , Structure-Activity Relationship , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/chemistry
12.
PLoS One ; 13(4): e0195542, 2018.
Article in English | MEDLINE | ID: mdl-29649293

ABSTRACT

One of the major drawbacks of many of the currently used cancer drugs are off-target effects. Targeted delivery is one method to minimize such unwanted and detrimental events. To actively target lung cancer cells, we have developed a conjugate of the apoptosis inducing protein cytochrome c with transferrin because the transferrin receptor is overexpressed by many rapidly dividing cancer cells. Cytochrome c and transferrin were cross-linked with a redox sensitive disulfide bond for the intra-cellular release of the protein upon endocytosis by the transferrin receptor. Confocal results demonstrated the cellular uptake of the cytochrome c-transferrin conjugate by transferrin receptor overexpressing A549 lung cancer cells. Localization studies further validated that this conjugate escaped the endosome. Additionally, an in vitro assay showed that the conjugate could induce apoptosis by activating caspase-3. The neo-conjugate not only maintained an IC50 value similar to the well known drug cisplatin (50 µM) in A549 cancer cells but also was nontoxic to the normal lung (MRC5) cells. Our neo-conjugate holds promise for future development to target cancers with enhanced transferrin receptor expression.


Subject(s)
Cytochromes c/metabolism , Drug Carriers/metabolism , Transferrin/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cytochromes c/chemistry , Cytochromes c/pharmacology , Cytochromes c/therapeutic use , Drug Carriers/chemistry , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Models, Molecular , Protein Conformation , Protein Transport , Receptors, Transferrin/metabolism , Transferrin/chemistry
13.
J Chem Educ ; 94(6): 722-725, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-29599566

ABSTRACT

Determining the catalytic activity of an enzyme can be the perfect method for its identification, for example during purification procedures or for isolation purposes. Herein, we used a pharmaceutically relevant protein to bring the concept of enzymatic activity to the classroom. We designed a hands-on interactive activity in which a medically relevant enzyme, asparaginase, was distinguished from a nonenzymatic protein based on its specific enzymatic activity. The experiment was carried out in the classroom, designed to impact different educational levels from elementary to high school. Our main purposes were to promote the emerging field of protein-based drugs as a source of scientific careers in bionanotechnology and to show the students an image of a "scientist" as that of a common and educated person working in an exciting profession. In addition of being inexpensive, this activity proved to be adaptable for various educational levels and can be easily implemented in different scenarios, for example, scientific fairs, some schools, and so forth.

14.
Mol Pharm ; 13(8): 2844-54, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27283751

ABSTRACT

Proteins often possess highly specific biological activities that make them potential therapeutics, but their physical and chemical instabilities during formulation, storage, and delivery have limited their medical use. Therefore, engineering of nanosized vehicles to stabilize protein therapeutics and to allow for targeted treatment of complex diseases, such as cancer, is of considerable interest. A micelle-like nanoparticle (NP) was designed for both, tumor targeting and stimulus-triggered release of the apoptotic protein cytochrome c (Cyt c). This system is composed of a Cyt c NP stabilized by a folate-receptor targeting amphiphilic copolymer (FA-PEG-PLGA) attached to Cyt c through a redox-sensitive bond. FA-PEG-PLGA-S-S-Cyt c NPs exhibited excellent stability under extracellular physiological conditions, whereas once in the intracellular reducing environment, Cyt c was released from the conjugate. Under the same conditions, the folate-decorated NP reduced folate receptor positive HeLa cell viability to 20%, while the same complex without FA only reduced it to 80%. Confocal microscopy showed that the FA-PEG-PLGA-S-S-Cyt c NPs were internalized by HeLa cells and were capable of endosomal escape. The specificity of the folate receptor-mediated internalization was confirmed by the lack of uptake by two folate receptor deficient cell lines: A549 and NIH-3T3. Finally, the potential as antitumor therapy of our folate-decorated Cyt c-based NPs was confirmed with an in vivo brain tumor model. In conclusion, we were able to create a stable, selective, and smart nanosized Cyt c delivery system.


Subject(s)
Cytochromes c/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , A549 Cells , Animals , Apoptosis , Cytochromes c/chemistry , Drug Carriers/chemistry , Drug Delivery Systems/methods , Glioma/metabolism , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Micelles , NIH 3T3 Cells , Polymers/chemistry
15.
J Nanomed Nanotechnol ; 6(3)2016 Jun.
Article in English | MEDLINE | ID: mdl-27088048

ABSTRACT

Photodynamic cancer therapy is still limited in its efficiency because of a lack of targeted methods avoiding non-specific toxicity. To overcome this we developed a system that is solely effective upon cellular uptake and intracellular activation by incorporating redox-sensitive chemistry. We used a nanoprecipitation method to obtain human serum albumin nanoparticles (HSA NP) with a diameter of 295 ± 5 nm and decorated them with the photosensitizer (PS) chlorin e6 (Ce6). The NP were stabilized using a redox-sensitive cross-linker to create a smart drug delivery system that is activated only upon NP disintegration in the reducing intracellular environment. Indeed, our drug delivery NP broke down in an environment emulating the reducing intracellular environment with 10 mM glutathione, but not under extracellular conditions. In contrast, the control cross-linked with glutaraldehyde did not break down in the reducing environment. Upon NP disintegration Ce6 fluorescence doubled as the result of diminished self-quenching. While the Ce6-HSA NP did not produce a significant amount of singlet oxygen upon irradiation, NP disintegration restored singlet oxygen production to about half of the value generated by the free Ce6. In vitro experiments with HeLa cells showed that the smart system was able to kill up to 81% of the cells while the glutaraldehyde cross-linked control only killed 56% of them at a drug concentration of 10 ng/ml. Also, Ce6 immobilization in HSA NP prevented dark toxicity in three different cell lines. For the first time, we demonstrate that it is possible to design a smart NP drug delivery system delivering a PS drug to cancer cells while avoiding toxicity prior to the uptake and irradiation. This finding may provide a means of designing more efficient PDT in cancer treatment.

17.
J Nanobiotechnology ; 13: 70, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26482026

ABSTRACT

BACKGROUND: This study provides fundamental information on the influence of graphene oxide (GO) nanosheets and glycans on protein catalytic activity, dynamics, and thermal stability. We provide evidence of protein stabilization by glycans and how this strategy could be implemented when GO nanosheets is used as protein immobilization matrix. A series of bioconjugates was constructed using two different strategies: adsorbing or covalently attaching native and glycosylated bilirubin oxidase (BOD) to GO. RESULTS: Bioconjugate formation was followed by FT-IR, zeta-potential, and X-ray photoelectron spectroscopy measurements. Enzyme kinetic parameters (k(m) and k(cat)) revealed that the substrate binding affinity was not affected by glycosylation and immobilization on GO, but the rate of enzyme catalysis was reduced. Structural analysis by circular dichroism showed that glycosylation did not affect the tertiary or the secondary structure of BOD. However, GO produced slight changes in the secondary structure. To shed light into the biophysical consequence of protein glycosylation and protein immobilization on GO nanosheets, we studied structural protein dynamical changes by FT-IR H/D exchange and thermal inactivation. CONCLUSIONS: It was found that glycosylation caused a reduction in structural dynamics that resulted in an increase in thermostability and a decrease in the catalytic activity for both, glycoconjugate and immobilized enzyme. These results establish the usefulness of chemical glycosylation to modulate protein structural dynamics and stability to develop a more stable GO-protein matrix.


Subject(s)
Biophysical Phenomena , Graphite/chemistry , Oxides/chemistry , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Biocatalysis , Caproates/chemistry , Circular Dichroism , Deuterium Exchange Measurement , Dextrans/chemistry , Glycosylation , Immobilized Proteins/metabolism , Kinetics , Nanoparticles/chemistry , Oxidation-Reduction , Photoelectron Spectroscopy , Spectroscopy, Fourier Transform Infrared , Static Electricity , Temperature
18.
BMC Biochem ; 16: 21, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26334686

ABSTRACT

BACKGROUND: Members of Rhizopus species are the most common cause of mucormycosis, a rare but often fatal fungal infection. Host induced pathogen apoptosis and pathogen induced host cell apoptosis are often involved in fungal infections. In many organisms, the release of mitochondrial cytochrome c can trigger apoptosis by activating caspase proteases, but the role of fungal cytochrome c in apoptosis remains unknown. RESULTS: DNA sequence encoding Rhizopus arrhizus cytochrome c was cloned and expressed in E. coli. Both native and recombinant cytochrome c were purified using ion exchange followed by gel filtration chromatography. The identities of purified proteins were confirmed by MALDI-MS and UV-Visible spectroscopy. For the first time, we demonstrated that Rhizopus arrhizus cytochrome c could activate human capspase-3 in HeLa cell extracts. We also found that Rhizopus arrhizus cytochrome c has redox potential, peroxidase activity, and spectral properties similar to human and horse cytochrome c proteins. CONCLUSIONS: Rhizopus arrhizus cytochrome c can activate human caspase-3 in HeLa cell extracts and it possesses similar physical and spectral properties as human and horse cytochrome c. This protein was found to have a previously unknown potential to activate human caspase-3, an important step in the apoptosis cascade.


Subject(s)
Caspase 3/metabolism , Cytochromes c/metabolism , Rhizopus/enzymology , Amino Acid Sequence , Animals , Enzyme Activation , HeLa Cells , Horses , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation
19.
FEBS Open Bio ; 5: 397-404, 2015.
Article in English | MEDLINE | ID: mdl-26101738

ABSTRACT

Lipid-protein complexes comprised of oleic acid (OA) non-covalently coupled to human/bovine α-lactalbumin, named HAMLET/BAMLET, display cytotoxic properties against cancer cells. However, there is still a substantial debate about the role of the protein in these complexes. To shed light into this, we obtained three different BAMLET complexes using varying synthesis conditions. Our data suggest that to form active BAMLET particles, OA has to reach critical micelle concentration with an approximate diameter of 250 nm. Proteolysis experiments on BAMLET show that OA protects the protein and is probably located on the surface, consistent with a micelle-like structure. Native or unfolded α-lactalbumin without OA lacked any tumoricidal activity. In contrast, OA alone killed cancer cells with the same efficiency at equimolar concentrations as its formulation as BAMLET. Our data show unequivocally that the cytotoxicity of the BAMLET complex is exclusively due to OA and that OA alone, when formulated as a micelle, is as toxic as the BAMLET complex. The contradictory literature results on the cytotoxicity of BAMLET might be explained by our finding that it was imperative to sonicate the samples to obtain toxic OA.

20.
J Nanomed Nanotechnol ; 6(5)2015 Oct.
Article in English | MEDLINE | ID: mdl-27182458

ABSTRACT

Effective cancer treatment needs both, passive and active targeting approaches, to achieve highly specific drug delivery to the target cells while avoiding cytotoxicity to normal cells. Protein drugs are useful in this context because they can display excellent specificity and potency. However, their use in therapeutic formulations is limited due to their physical and chemical instability during storage and administration. Polysaccharides have been used to stabilize proteins during formulation and delivery. To accomplish both, stabilization and targeting simultaneously, the apoptosis-inducing protein cytochrome c (Cyt c) was modified with the polysaccharide hyaluronic acid (HA) because its corresponding receptor CD44 is overexpressed in many cancers. Cyt c-HA bioconjugates were formed using low and high molecular weight HA (8 kDa and 1 MDa) with a resultant Cyt c loading percentage of 4%. Circular dichroism and a cell-free caspase assay showed minor structural changes and high bioactivity (more than 80% caspase activation) of Cyt c, respectively, after bioconjugate formation. Two CD44-positive cancer cells lines, HeLa and A549 cells, and two CD44-negative normal cell lines, Huvec and NIH-3T3 cells, were incubated with the samples to assess selectivity and cytotoxicity. After 24 h of incubation with the samples, cancer cell viability was reduced at least 3-fold while CD44-negative control cell lines remained minimally affected. Fluorescence imaging confirmed selective internalization of the Cyt c-HA construct by CD44-positive cancer cell lines. These results demonstrate the development of a drug delivery system that incorporates passive and active targeting which is essential for cancer treatment.

SELECTION OF CITATIONS
SEARCH DETAIL
...