Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Extracell Vesicles ; 12(7): e12332, 2023 07.
Article in English | MEDLINE | ID: mdl-37353884

ABSTRACT

The release of growth factors, cytokines and extracellular matrix modifiers by activated platelets is an important step in the process of healthy wound healing. Extracellular vesicles (EVs) released by activated platelets carry this bioactive cargo in an enriched form, and may therefore represent a potential therapeutic for the treatment of delayed wound healing, such as chronic wounds. While EVs show great promise in regenerative medicine, their production at clinical scale remains a critical challenge and their tolerability in humans is still to be fully established. In this work, we demonstrate that Ligand-based Exosome Affinity Purification (LEAP) chromatography can successfully isolate platelet EVs (pEVs) of clinical grade from activated platelets, which retain the regenerative properties of the parent cell. LEAP-isolated pEVs display the expected biophysical features of EV populations and transport essential proteins in wound healing processes, including insulin growth factor (IGF) and transforming growth factor beta (TGF-ß). In vitro studies show that pEVs induce proliferation and migration of dermal fibroblasts and increase dermal endothelial cells' angiogenic potential, demonstrating their wound healing potential. pEV treatment activates the ERK and Akt signalling pathways within recipient cells. In a first-in-human, double-blind, placebo-controlled, phase I clinical trial of healthy volunteer adults, designed primarily to assess safety in the context of wound healing, we demonstrate that injections of LEAP-purified pEVs in formulation buffer are safe and well tolerated (Plexoval II study, ACTRN12620000944932). As a secondary objective, biological activity in the context of wound healing rate was assessed. In this cohort of healthy participants, in which the wound bed would not be expected to be deficient in the bioactive cargo that pEVs carry, all wounds healed rapidly and completely and no difference in time to wound closure of the treated and untreated wounds was observed at the single dose tested. The outcomes of this study evidence that pEVs manufactured through the LEAP process can be injected safely in humans as a potential wound healing treatment, and warrant further study in clinical trials designed expressly to assess therapeutic efficacy in patients with delayed or disrupted wound healing.


Subject(s)
Extracellular Vesicles , Hematopoietic Stem Cell Transplantation , Adult , Humans , Blood Platelets/metabolism , Endothelial Cells , Extracellular Vesicles/metabolism , Wound Healing/physiology
2.
Pharmaceutics ; 15(5)2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37242631

ABSTRACT

Despite the clinical benefits that chemotherapeutics has had on the treatment of breast cancer, drug resistance remains one of the main obstacles to curative cancer therapy. Nanomedicines allow therapeutics to be more targeted and effective, resulting in enhanced treatment success, reduced side effects, and the possibility of minimising drug resistance by the co-delivery of therapeutic agents. Porous silicon nanoparticles (pSiNPs) have been established as efficient vectors for drug delivery. Their high surface area makes them an ideal carrier for the administration of multiple therapeutics, providing the means to apply multiple attacks to the tumour. Moreover, immobilising targeting ligands on the pSiNP surface helps direct them selectively to cancer cells, thereby reducing harm to normal tissues. Here, we engineered breast cancer-targeted pSiNPs co-loaded with an anticancer drug and gold nanoclusters (AuNCs). AuNCs have the capacity to induce hyperthermia when exposed to a radiofrequency field. Using monolayer and 3D cell cultures, we demonstrate that the cell-killing efficacy of combined hyperthermia and chemotherapy via targeted pSiNPs is 1.5-fold higher than applying monotherapy and 3.5-fold higher compared to using a nontargeted system with combined therapeutics. The results not only demonstrate targeted pSiNPs as a successful nanocarrier for combination therapy but also confirm it as a versatile platform with the potential to be used for personalised medicine.

3.
ACS Appl Mater Interfaces ; 14(49): 54539-54549, 2022 Dec 14.
Article in English | MEDLINE | ID: mdl-36469497

ABSTRACT

An approach to differentially modify the internal surface of porous silicon nanoparticles (pSiNPs) with hydrophobic dodecene and the external surface with antifouling poly-N-(2-hydroxypropyl) acrylamide (polyHPAm) as well as a cell-targeting peptide was developed. Specifically, to generate these core-shell pSiNPs, the interior surface of a porous silicon (pSi) film was hydrosilylated with 1-dodecene, followed by ultrasonication to create pSiNPs. The new external surfaces were modified by silanization with a polymerization initiator, and surface-initiated atom transfer radical polymerization was performed to introduce polyHPAm brushes. Afterward, a fraction of the polymer side chain hydroxyl groups was activated to conjugate cRGDfK─a peptide with a high affinity and selectivity for the ανß3 integrin receptor that is overexpressed in prostate and melanoma cancers. Finally, camptothecin, a hydrophobic anti-cancer drug, was successfully loaded into the pores. This drug delivery system showed excellent colloidal stability in a cell culture medium, and the in vitro drug release kinetics could be fine-tuned by the combination of internal and external surface modifications. In vitro studies by confocal microscopy and flow cytometry revealed improved cellular association attributed to cRGDfK. Furthermore, the cell viability results showed that the drug-loaded and peptide-functionalized nanoparticles had enhanced cytotoxicity toward a C4-2B prostate carcinoma cell line in both 2D cell culture and a 3D spheroid model.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Humans , Silicon/chemistry , Porosity , Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Cell Line, Tumor
4.
J Nanobiotechnology ; 20(1): 482, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36384747

ABSTRACT

Inspired by nature, green chemistry uses various biomolecules, such as proteins, as reducing agents to synthesize metallic nanostructures. This methodology provides an alternative route to conventional harsh synthetic processes, which include polluting chemicals. Tuning the resulting nanostructure properties, such as their size and shape, is challenging as the exact mechanism involved in their formation is still not well understood. This work reports a well-controlled method to program gold nanostructures' shape, size, and aggregation state using only one protein type, mucin, as a reduction and capping material in a one-pot bio-assisted reaction. Using mucin as a gold reduction template while varying its tertiary structure via the pH of the synthesis, we demonstrate that spherical, coral-shaped, and hexagonal gold crystals can be obtained and that the size can be tuned over three orders of magnitude. This is achieved by leveraging the protein's intrinsic reducing properties and pH-induced conformational changes. The systematic study of the reaction kinetics and growth steps developed here provides an understanding of the mechanism behind this phenomenon. We further show that the prepared gold nanostructures exhibit tunable photothermal properties that can be optimized for various hyperthermia-induced antibacterial applications.


Subject(s)
Metal Nanoparticles , Nanostructures , Gold/chemistry , Metal Nanoparticles/chemistry , Mucins
5.
ACS Appl Mater Interfaces ; 13(42): 49581-49588, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34636533

ABSTRACT

Quantum-sized gold nanoclusters (AuNCs) are emerging as theranostic agents-those that combine diagnostics and therapeutic properties-given their ultrasmall size <3 nm, which makes them behave more like a molecule rather than a nanoparticle. This molecule-like behavior endows AuNCs with interesting properties including photoluminescence, catalytic activity, and paramagnetism-all without the presence of any toxic heavy metal. But despite these fundamental advances, scalable synthetic approaches to produce high-quality AuNCs with well-controlled and programmable properties for biological applications as well as methods to determine their structure-property relationships are not widely available. In this Perspective, we will discuss what is known so far about AuNCs as well as how to move forward to propel AuNCs as a theranostic agent of choice for many biomedical applications.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Theranostic Nanomedicine , Biomedical Research , Humans , Materials Testing , Particle Size
6.
Cancer Res ; 81(7): 1704-1718, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33547161

ABSTRACT

The androgen receptor (AR) is the key oncogenic driver of prostate cancer, and despite implementation of novel AR targeting therapies, outcomes for metastatic disease remain dismal. There is an urgent need to better understand androgen-regulated cellular processes to more effectively target the AR dependence of prostate cancer cells through new therapeutic vulnerabilities. Transcriptomic studies have consistently identified lipid metabolism as a hallmark of enhanced AR signaling in prostate cancer, yet the relationship between AR and the lipidome remains undefined. Using mass spectrometry-based lipidomics, this study reveals increased fatty acyl chain length in phospholipids from prostate cancer cells and patient-derived explants as one of the most striking androgen-regulated changes to lipid metabolism. Potent and direct AR-mediated induction of ELOVL fatty acid elongase 5 (ELOVL5), an enzyme that catalyzes fatty acid elongation, was demonstrated in prostate cancer cells, xenografts, and clinical tumors. Assessment of mRNA and protein in large-scale data sets revealed ELOVL5 as the predominant ELOVL expressed and upregulated in prostate cancer compared with nonmalignant prostate. ELOVL5 depletion markedly altered mitochondrial morphology and function, leading to excess generation of reactive oxygen species and resulting in suppression of prostate cancer cell proliferation, 3D growth, and in vivo tumor growth and metastasis. Supplementation with the monounsaturated fatty acid cis-vaccenic acid, a direct product of ELOVL5 elongation, reversed the oxidative stress and associated cell proliferation and migration effects of ELOVL5 knockdown. Collectively, these results identify lipid elongation as a protumorigenic metabolic pathway in prostate cancer that is androgen-regulated, critical for metastasis, and targetable via ELOVL5. SIGNIFICANCE: This study identifies phospholipid elongation as a new metabolic target of androgen action that is critical for prostate tumor metastasis.


Subject(s)
Fatty Acid Elongases/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , RNA, Small Interfering/therapeutic use , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Fatty Acid Elongases/genetics , Fatty Acid Elongases/physiology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/pharmacology , Receptors, Androgen/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Nano Lett ; 21(1): 476-484, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33350838

ABSTRACT

We introduce xanthate-functionalized poly(cyclic imino ethers)s (PCIEs), specifically poly(2-ethyl-2-oxazoline) and poly(2-ethyl-2-oxazine) given their stealth characteristics, as an attractive alternative to conventional thiol-based ligands for the synthesis of highly monodisperse and fluorescent gold nanoclusters (AuNCs). The xanthate in the PCIEs interacts with Au ions, acting as a well-controlled template for the direct formation of PCIE-AuNCs. This method yields red-emitting AuNCs with a narrow emission peak (λem = 645 nm), good quantum yield (4.3-4.8%), long fluorescence decay time (∼722-844 ns), and unprecedented product yield (>98%). The PCIE-AuNCs exhibit long-term colloidal stability, biocompatibility, and antifouling properties, enabling a prolonged blood circulation, lower nonspecific accumulation in major organs, and better renal clearance when compared with AuNCs without polymer coating. The advances made here in the synthesis of metal nanoclusters using xanthate-functionalized PCIEs could propel the production of highly monodisperse, biocompatible, and renally clearable nanoprobes in large-scale for different theranostic applications.

8.
Nat Nanotechnol ; 16(1): 37-46, 2021 01.
Article in English | MEDLINE | ID: mdl-33349685

ABSTRACT

Induced tolerogenic dendritic cells are a powerful immunotherapy for autoimmune disease that have shown promise in laboratory models of disease and early clinical trials. In contrast to conventional immunosuppressive treatments, tolerogenic immunotherapy leverages the cells and function of the immune system to quell the autoreactive lymphocytes responsible for damage and disease. The principle techniques of isolating and reprogramming dendritic cells (DCs), central to this approach, are well characterized. However, the broader application of this technology is limited by its high cost and bespoke nature. Nanomedicine offers an alternative route by performing this reprogramming process in situ. Here, we review the challenges and opportunities in using nanoparticles as a delivery mechanism to target DCs and induce immunomodulation, emphasizing their versatility. We then highlight their potential to solve critical problems in organ transplantation and increasingly prevalent autoimmune disorders such as type 1 diabetes mellitus and multiple sclerosis, where new immunotherapy approaches have begun to show promise.


Subject(s)
Dendritic Cells/immunology , Drug Delivery Systems/methods , Nanomedicine/methods , Nanoparticles , Transplantation Tolerance/immunology , Animals , Antigens/immunology , Autoimmune Diseases/therapy , Humans , Immunomodulation , Immunosuppressive Agents/administration & dosage , Nanoparticles/chemistry , Particle Size
9.
Biomater Sci ; 9(1): 133-147, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33135714

ABSTRACT

Targeted delivery of chemotherapeutics to cancer cells has the potential to yield high drug concentrations in cancer cells while minimizing any unwanted side effects. However, the development of multidrug resistance in cancer cells may impede the accumulation of chemotherapy drugs within these, decreasing its therapeutic efficacy. Downregulation of multidrug resistance-related proteins such as MRP1 with small interfering RNA (siRNA) is a promising approach in the reversal of drug resistance. The co-delivery of doxorubicin (Dox) and siRNA against MRP1 (siMRP1) by using nanoparticles comprised of biocompatible porous silicon (pSi) presents itself as a novel opportunity to utilize the biomaterial's high loading capacity and large accessible surface area. Additionally, to increase the selectivity and retention of the delivery vehicle at the tumor site, nanobodies were incorporated onto the nanoparticle surface via a polyethylene glycol (PEG) linker directed towards either the epidermal growth factor receptor (EGFR) or the prostate specific membrane antigen (PSMA). The nanobody-displaying pSi nanoparticles (pSiNPs) demonstrated effective gene silencing, inhibiting MRP1 expression by 74 ± 6% and 74 ± 4% when incubated with EGFR-pSiNPs and PSMA-pSiNPs, respectively, in prostate cancer cells. The downregulation of MRP1 led to a further increase in cytotoxicity when both siRNA and Dox were delivered in conjunction in both cancer cell monocultures and spheroids when compared to free Dox or Dox and a scrambled sequence of siRNA. Altogether, nanobody-displaying pSiNPs are an effective carrier for the dual delivery of both siRNA and Dox for cancer treatment.


Subject(s)
Nanoparticles , Silicon , Cell Line, Tumor , Doxorubicin , Drug Delivery Systems , Male , Porosity , RNA, Small Interfering
10.
Adv Healthc Mater ; 10(6): e2001594, 2021 03.
Article in English | MEDLINE | ID: mdl-33274851

ABSTRACT

Over the last thirty years, research in nanomedicine has widely been focused on applications in cancer therapeutics. However, despite the plethora of reported nanoscale drug delivery systems that can successfully eradicate solid tumor xenografts in vivo, many of these formulations have not yet achieved clinical translation. This issue particularly pertains to the delivery of small interfering RNA (siRNA), a highly attractive tool for selective gene targeting. One of the likely reasons behind the lack of translation is that current in vivo models fail to recapitulate critical elements of clinical solid tumors that may influence drug response, such as cellular heterogeneity in the tumor microenvironment. This study incorporates a more clinically relevant model for assessing siRNA delivery systems; ex vivo culture of prostate cancer harvested from patients who have undergone radical prostatectomy, denoted patient-derived explants (PDE). The model retains native human tissue architecture, microenvironment, and cell signaling pathways. Porous silicon nanoparticles (pSiNPs) behavior in this model is investigated and compared with commonly used 3D cancer cell spheroids for their efficacy in the delivery of siRNA directed against the androgen receptor (AR), a key driver of prostate cancer.


Subject(s)
Nanoparticles , Prostatic Neoplasms , Cell Line, Tumor , Humans , Male , Nanomedicine , Prostatic Neoplasms/therapy , RNA, Small Interfering , Tumor Microenvironment
11.
Biomaterials ; 240: 119791, 2020 05.
Article in English | MEDLINE | ID: mdl-32109589

ABSTRACT

In advanced breast cancer (BCa) patients, not the primary tumor, but the development of distant metastases, which occur mainly in the organ bone, and their adverse health effects are responsible for high mortality. Targeted delivery of already known drugs which displayed potency, but rather unfavorable pharmacokinetic properties, might be a promising approach to overcome the current limitations of metastatic BCa therapy. Camptothecin (CPT) is a highly cytotoxic chemotherapeutic compound, yet poorly water-soluble and non-specific. Here, CPT was loaded into porous silicon nanoparticles (pSiNP) displaying the epidermal growth factor receptor (EGFR)-targeting antibody (Ab) cetuximab to generate a soluble and targeted nanoscale delivery vehicle for cancer treatment. After confirming the cytotoxic effect of targeted CPT-loaded pSiNP in vitro on MDA-MB-231BO cells, nanoparticles were studied in a humanized BCa bone metastasis mouse model. Humanized tissue-engineered bone constructs (hTEBCs) provided a humanized microenvironment for BCa bone metastases in female NOD-scid IL2Rgnull (NSG) mice. Actively targeted CPT-loaded pSiNP led to a reduction of orthotopic primary tumor growth, increased survival rate and significant decrease in hTEBC and murine lung, liver and bone metastases. This study demonstrates that targeted delivery via pSiNP is an effective approach to employ CPT and other potent anti-cancer compounds with poor pharmacokinetic profiles in cancer therapy.


Subject(s)
Breast Neoplasms , Nanoparticles , Animals , Breast Neoplasms/drug therapy , Camptothecin , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Silicon , Tumor Microenvironment
12.
ACS Appl Mater Interfaces ; 11(26): 22993-23005, 2019 Jul 03.
Article in English | MEDLINE | ID: mdl-31252458

ABSTRACT

Gene silencing by RNA interference is a powerful technology with broad applications. However, this technology has been hampered by the instability of small interfering RNA (siRNA) molecules in physiological conditions and their inefficient delivery into the cytoplasm of target cells. Porous silicon nanoparticles have emerged as a potential delivery vehicle to overcome these limitations-being able to encapsulate RNA molecules within the porous matrix and protect them from degradation. Here, key variables were investigated that influence siRNA loading into porous silicon nanoparticles. The effect of modifying the surface of porous silicon nanoparticles with various amino-functional molecules as well as the effects of salt and chaotropic agents in facilitating siRNA loading was examined. Maximum siRNA loading of 413 µg/(mg of porous silicon nanoparticles) was found when the nanoparticles were modified by a fourth generation polyamidoamine dendrimer. Low concentrations of urea or salt increased loading capacity: an increase in RNA loading by 19% at a concentration of 0.05 M NaCl or 21% at a concentration of 0.25 M urea was observed when compared to loading in water. Lastly, it was demonstrated that dendrimer-functionalized nanocarriers are able to deliver siRNA against ELOVL5, a target for the treatment of advanced prostate cancer.


Subject(s)
Gene Silencing , Gene Transfer Techniques , Nanoparticles/chemistry , RNA, Small Interfering/chemistry , Dendrimers/chemistry , Dendrimers/pharmacology , Fatty Acid Elongases/antagonists & inhibitors , Fatty Acid Elongases/genetics , Humans , Male , Nanoparticles/therapeutic use , Polyamines/chemistry , Polyamines/pharmacology , Porosity , Prostatic Neoplasms/drug therapy , RNA Interference , RNA, Small Interfering/genetics , Silicon/chemistry
14.
ACS Appl Mater Interfaces ; 9(49): 42601-42611, 2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29154535

ABSTRACT

Continuing our research efforts in developing mesoporous silicon nanoparticle-based biomaterials for cancer therapy, we employed here porous silicon nanoparticles as a nanocarrier to deliver contrast agents to diseased cells. Nanoconfinement of small molecule Gd-chelates (L1-Gd) enhanced the T1 contrast dramatically compared to distinct Gd-chelate (L1-Gd) by virtue of its slow tumbling rate, increased number of bound water molecules, and their occupancy time. The newly synthesized Gd-chelate (L1-Gd) was covalently grafted on silicon nanostructures and conjugated to an antibody specific for epidermal growth factor receptor (EGFR) via a hydrazone linkage. The salient feature of this nanosized contrast agent is the capability of EGFR targeted delivery to cancer cells. Mesoporous silicon nanoparticles were chosen as the nanocarrier because of their high porosity, high surface area, and excellent biodegradability. This type of nanosized contrast agent also performs well in high magnetic fields.


Subject(s)
Nanoparticles , Contrast Media , ErbB Receptors , Gadolinium , Magnetic Resonance Imaging , Silicon
15.
ACS Appl Mater Interfaces ; 9(47): 41159-41167, 2017 Nov 29.
Article in English | MEDLINE | ID: mdl-29116739

ABSTRACT

Gold nanoclusters (Au NCs) have become a promising nanomaterial for cancer therapy because of their biocompatibility and fluorescent properties. In this study, the effect of ultrasmall protein-stabilized 2 nm Au NCs on six types of mammalian cells (fibroblasts, B-lymphocytes, glioblastoma, neuroblastoma, and two types of prostate cancer cells) under electromagnetic radiation is investigated. Cellular association of Au NCs in vitro is concentration-dependent, and Au NCs have low intrinsic toxicity. However, when Au NC-incubated cells are exposed to a 1 GHz electromagnetic field (microwave radiation), cell viability significantly decreases, thus demonstrating that Au NCs exhibit specific microwave-dependent cytotoxicity, likely resulting from localized heating. Upon i.v. injection in mice, Au NCs are still present at 24 h post administration. Considering the specific microwave-dependent cytotoxicity and low intrinsic toxicity, our work suggests the potential of Au NCs as effective and safe nanomedicines for cancer therapy.


Subject(s)
Nanostructures , Animals , Cell Survival , Electromagnetic Radiation , Gold , Metal Nanoparticles , Mice
16.
ACS Appl Mater Interfaces ; 9(39): 33707-33716, 2017 Oct 04.
Article in English | MEDLINE | ID: mdl-28910076

ABSTRACT

In order to address the issue of pathogenic bacterial colonization of diabetic wounds, a more direct and robust approach is required, which relies on a physical form of bacterial destruction in addition to the conventional biochemical approach (i.e., antibiotics). Targeted bacterial destruction through the use of photothermally active nanomaterials has recently come into the spotlight as a viable approach to solving the rising problem of antibiotic resistant microorganisms. Materials with high absorption coefficients in the near-infrared (NIR) region of the electromagnetic spectrum show promise as alternative antibacterial therapeutic agents, since they preclude the development of bacterial resistance and can be activated on demand. Here were report on a novel approach for the fabrication of gold nanoparticle decorated porous silicon nanopillars with tunable geometry that demonstrate excellent photothermal conversion properties when irradiated with a 808 nm laser. These photothermal antibacterial properties are demonstrated in vitro against the Gram-positive bacteria Staphylococcus aureus (S. aureus) and Gram-negative Escherichia coli (E. coli). Results show a reduction in bacterial viability of up to 99% after 10 min of laser irradiation. We also show an increase in antibacterial performance after modifying the nanopillars with S. aureus targeting antibodies causing up to a 10-fold increase in bactericidal efficiency compared to E. coli. In contrast, the nanomaterial resulted in minimal disruption of metabolic processes in human foreskin fibroblasts (HFF) after an equivalent period of irradiation.


Subject(s)
Nanostructures , Anti-Bacterial Agents , Bacterial Infections , Escherichia coli , Gold , Humans , Metal Nanoparticles , Microbial Sensitivity Tests , Porosity , Silicon , Staphylococcus aureus
17.
Small ; 13(29)2017 08.
Article in English | MEDLINE | ID: mdl-28570785

ABSTRACT

There is a pressing need to develop more effective therapeutics to fight cancer. An idyllic chemotherapeutic is expected to overcome drug resistance of tumors and minimize harmful side effects to healthy tissues. Antibody-functionalized porous silicon nanoparticles loaded with a combination of chemotherapy drug and gold nanoclusters (AuNCs) are developed. These nanocarriers are observed to selectively deliver both payloads, the chemotherapy drug and AuNCs, to human B cells. The accumulation of AuNCs to target cells and subsequent exposure to an external electromagnetic field in the microwave region render them more susceptible to the codelivered drug. This approach represents a targeted two-stage delivery nanocarrier that benefits from a dual therapeutic action that results in enhanced cytotoxicity.


Subject(s)
Gold/chemistry , Nanoparticles/chemistry , Neoplasms/therapy , Silicon/chemistry , Drug Delivery Systems/methods , Porosity
18.
ACS Appl Mater Interfaces ; 9(13): 11461-11471, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28299925

ABSTRACT

Gene therapy has arisen as a pioneering technique to treat diseases by direct employment of nucleic acids as medicine. The major historical problem is to develop efficient and safe systems for the delivery of therapeutic genes into the target cells. Carbon nanotubes (CNTs) have demonstrated considerable promise as delivery vectors due to their (i) high aspect ratio and (ii) capacity to translocate through plasma membranes, known as the nanoneedle effect. To leverage these advantages, close attention needs to be paid to the physicochemical characteristics of the CNTs used. CNTs with different diameters (thinner and thicker) were treated by chemical oxidation to produce shorter fragments. Rigid (thick) and flexible (thin) CNTs, and their shortened versions, were coated with polyallylamine (ppAA) by plasma-enhanced chemical vapor deposition. The ppAA coating leads to a positively charged CNT surface that is able to electrostatically bind the green fluorescent protein plasmid reporter. This study shows how rigidity and length can affect their (i) behavior in biological media, (ii) ability to transfect in vitro, and (iii) biodistribution in vivo. This study also generates a set of basic design rules for the development of more efficient CNT-based gene-delivery vectors.


Subject(s)
Nanotubes, Carbon , Gene Transfer Techniques , Genetic Therapy , Plasmids , Tissue Distribution
19.
ACS Appl Mater Interfaces ; 7(50): 27755-64, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26629977

ABSTRACT

We demonstrate microwave-induced heating of gold nanoparticles and nanorods. An appreciably higher and concentration-dependent microwave-induced heating rate was observed with aqueous dispersions of the nanomaterials as opposed to pure water and other controls. Grafted with the thermoresponsive polymer poly(N-isopropylacrylamide), these gold nanomaterials react to microwave-induced heating with a conformational change in the polymer shell, leading to particle aggregation. We subsequently covalently immobilize concanavalin A (Con A) on the thermoresponsive gold nanoparticles. Con A is a bioreceptor commonly used in bacterial sensors because of its affinity for carbohydrates on bacterial cell surfaces. The microwave-induced thermal transitions of the polymer reversibly switch on and off the display of Con A on the particle surface and hence the interactions of the nanomaterials with carbohydrate-functionalized surfaces. This effect was determined using linear sweep voltammetry on a methyl-α-d-mannopyranoside-functionalized electrode.


Subject(s)
Acrylic Resins/chemistry , Concanavalin A/chemistry , Metal Nanoparticles/chemistry , Acrylic Resins/chemical synthesis , Concanavalin A/chemical synthesis , Gold/chemistry , Microwaves , Particle Size , Surface Properties , Temperature
20.
ACS Nano ; 7(11): 10066-74, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24128271

ABSTRACT

We manipulate the passive release rates of DNA payloads on protein coronas formed around nanoparticles (NPs) by varying the corona composition. The coronas are prepared using a mixture of hard and soft corona proteins. We form coronas around gold nanorods (NRs), nanobones (NBs), and carbon nanotubes (CNTs) from human serum (HS) and find that tuning the amount of human serum albumin (HSA) in the NR-coronas (NR-HS-DNA) changes the payload release profile. The effect of buffer strength, HS concentration, and concentration of the cetyltrimethylammonium bromide (CTAB) passivating the NP surfaces on passive release is explored. We find that corona properties play an important role in passive release, and concentrations of CTAB, HS, and phosphate buffer used in corona formation can tune payload release profiles. These advances in understanding protein corona properties bring us closer toward developing a set of basic design rules that enable their manipulation and optimization for particular biological applications.


Subject(s)
DNA/chemistry , Nanoparticles/chemistry , Proteins/chemistry , Adsorption , DNA/analysis , Drug Carriers , Gold/chemistry , Humans , Ligands , Metal Nanoparticles , Nanotechnology/methods , Nanotubes, Carbon/chemistry , Particle Size , Serum Albumin/chemistry , Spectrometry, Fluorescence , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...