Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Cancer Gene Ther ; 31(3): 464-471, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38177307

ABSTRACT

KRAS mutations occur commonly in the lung and can lead to the development of non-small cell lung cancer (NSCLC). While the mutated KRAS protein is a neoantigen, it usually does not generate an effective anti-tumor immune response on mucosal/epithelial surfaces. Despite this, mutated KRAS remains a potential target for immunotherapy since immune targeting of this protein in animal models has been effective at eliminating tumor cells. We attempted to develop a KRAS vaccine using mutated and wild-type KRAS peptides in combination with a nanoemulsion (NE) adjuvant. The efficacy of this approach was tested in an inducible mutant KRAS-mouse lung tumor model. Animals were immunized intranasally using NE with KRAS peptides. These animals had decreased CD4+FoxP3+ T cells in both lymph nodes and spleen. Immunized animals also showed higher IFN-γ and IL-17a levels to mutated KRAS that were produced by CD8+ T cells and enhancement in KRAS-specific Th1 and Th17 responses that persisted for 3 months after the last vaccination. Importantly, the immunized animals had significantly decreased tumor incidence compared to control animals. In conclusion, a mucosal approach to KRAS vaccination demonstrated the ability to induce local KRAS-specific immune responses in the lung and resulted in reduced tumor incidence.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/genetics , Lung Neoplasms/prevention & control , Protein Subunit Vaccines , Proto-Oncogene Proteins p21(ras)/genetics , CD8-Positive T-Lymphocytes , Disease Models, Animal , Peptides/genetics , Mutation
2.
Pharmaceutics ; 15(7)2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37514184

ABSTRACT

The objective of the study was to explore the feasibility of a new drug delivery system using laponite (LAP) and cyclic poly(ethylene glycol) (cPEG). Variously shaped and flexible hybrid nanocrystals were made by both the covalent and physical attachment of chemically homogeneous cyclized PEG to laponite nanodisc plates. The size of the resulting, nearly spherical particles ranged from 1 to 1.5 µm, while PEGylation with linear methoxy poly (ethylene glycol) (mPEG) resulted in fragile sheets of different shapes and sizes. When infused with 10% doxorubicin (DOX), a drug commonly used in the treatment of various cancers, the LAP-cPEG/DOX formulation was transparent and maintained liquid-like homogeneity without delamination, and the drug loading efficiency of the LAP-cPEG nano system was found to be higher than that of the laponite-poly(ethylene glycol) LAP-mPEG system. Furthermore, the LAP-cPEG/DOX formulation showed relative stability in phosphate-buffered saline (PBS) with only 15% of the drug released. However, in the presence of human plasma, about 90% of the drug was released continuously over a period of 24 h for the LAP-cPEG/DOX, while the LAP-mPEG/DOX formulation released 90% of DOX in a 6 h burst. The results of the cell viability assay indicated that the LAP-cPEG/DOX formulation could effectively inhibit the proliferation of A549 lung carcinoma epithelial cells. With the DOX concentration in the range of 1-2 µM in the LAP-cPEG/DOX formulation, enhanced drug effects in both A549 lung carcinoma epithelial cells and primary lung epithelial cells were observed compared to LAP-mPEG/DOX. The unique properties and effects of cPEG nanoparticles provide a potentially better drug delivery system and generate interest for further targeting studies and applications.

3.
Microbiol Spectr ; 10(2): e0250621, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35412373

ABSTRACT

Wound infections with methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE) are particularly difficult to treat and present a great challenge to clinicians. Nanoemulsions (NE) are novel oil-in-water emulsions formulated from soybean oil, water, solvent, and surfactants such as benzalkonium chloride (BZK). An optimal ratio of those components produces nanometer-sized particles with the positive-charged surfactant at their oil-water interface. We sought to investigate antimicrobial NE as a novel treatment to address wounds co-infected by MRSA and VRE. Swine split-thickness skin wounds were first infected with MRSA and/or VRE, then treated with the nanoemulsion formulation (X-1735) or placebo controls. Bacterial viability after treatment were determined by nutrient agar plates for total, MRSA-specific, and VRE-specific loads. In addition, inflammation indexes were scored by histopathology. When VRE infected wounds were treated with X-1735, they contained 103 lower VRE CFU counts across a 2-week period compared with placebo. Once co-infected MRSA and VRE split-thickness wounds were successfully established, topical treatment of co-infected wounds with X-1735 resulted in a reduction of bacteria by 2 to 3 logs (compared with placebo) at 3- and 14-day postinfection time points. Importantly, X-1735 was effective in significantly alleviating multilevel inflammation in the treated wounds. X-1735 is a new antimicrobial that is safe to apply to open wounds and effectively kills MRSA and VRE. It appears to also reduce inflammation in these co-infected wounds. The data suggest that this approach offers promise as an antimicrobial for open wounds with MRSA and VRE co-infection. IMPORTANCE Infections, specifically polymicrobial, can cause serious consequences when it comes to wound treatment. Prolonged treatment with antibiotics can lead to an increased risk of bacterial resistance; co-infections can complicate treatment options even further. Our research proposes a novel nanoemulsion treatment for two of the most common antibiotic resistant bacteria: methicillin-resistant Staphylococcus aureus (MRSA) and Vancomycin-resistant enterococci (VRE). This optimized topical treatment formulation not only significantly reduces inflammation and infection in MRSA or VRE infected wounds, but also in MRSA and VRE co-infected wounds as well. The work aims to provide an alternative treatment approach for multidrug-resistant organisms and decrease dependence on systemic treatments.


Subject(s)
Gram-Positive Bacterial Infections , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Vancomycin-Resistant Enterococci , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial , Gram-Positive Bacterial Infections/microbiology , Inflammation , Staphylococcal Infections/microbiology , Swine , Water
4.
Drug Deliv ; 28(1): 2150-2159, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34617850

ABSTRACT

We examined formulating a new antifungal agent, posaconazole (POS) and its derivatives, with different molecular vehicles. Several combinations of drug and carrier molecules were synthesized, and their antifungal activities were evaluated against Aspergillus fumigatus. Posaconazole and four of its derivatives were conjugated to either generation 5 (G5) dendrimers or partially modified G5 dendrimers. The in vitro antifungal activities of these compounds suggest that conjugates with specific chemical linkages showed better fungistatic activity than direct conjugates to POS. In particular, a polyethylene glycol (PEG)-imidazole modified G5 dendrimer demonstrated improved antifungal efficacy relative to the parent G5 molecule. Further studies were then conducted with POS derived molecules coupled to PEG-imidazole modified G5 dendrimers to achieve a highly soluble and active conjugate of POS. This conjugated macromolecule averaged 23 POS molecules per G5 and had a high solubility with 50 mg/mL, which improved the molar solubility of POS from less than 0.03 mg/mL to as high as 16 mg/mL in water. The primary release profile of the drug in human plasma was extended to over 72 h, which is reflected in the in vitro inhibition of A. fumigatus growth of over 96 h. These POS-polymer conjugates appear to be novel and efficient antifungal agents.


Subject(s)
Antifungal Agents/administration & dosage , Aspergillus fumigatus/drug effects , Dendrimers/chemistry , Nanoparticles/chemistry , Triazoles/administration & dosage , Antifungal Agents/pharmacology , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Liberation , Imidazoles/pharmacology , Polyethylene Glycols/chemistry , Triazoles/pharmacology
5.
J Nanobiotechnology ; 18(1): 135, 2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32948179

ABSTRACT

BACKGROUND: Targeted contrast nanoparticles for breast tumor imaging facilitates early detection and improves treatment efficacy of breast cancer. This manuscript reports the development of an epidermal growth factor receptor-2 (HER-2) specific, bi-modal, dendrimer conjugate to enhance computed tomography (CT) and magnetic resonance imaging (MRI) of HER-2-positive breast cancer. This material employs generation 5 poly(amidoamine) dendrimers, encapsulated gold nanoparticles, chelated gadolinium, and anti-human HER-2 antibody to produce the nanoparticle contrast agent. RESULTS: Testing in two mouse tumor models confirms this contrast agent's ability to image HER-2 positive tumors. Intravenous injection of this nanoparticle in mice bearing HER-2 positive mammary tumors significantly enhances MRI signal intensity by ~ 20% and improves CT resolution and contrast by two-fold. Results by flow cytometry and confocal microscopy validate the specific targeting of the conjugate and its internalization in human HER-2 positive cells. CONCLUSION: These results demonstrate that this nanoparticle conjugate can efficiently target and image HER-2 positive tumors in vivo and provide a basis for the development of this diagnostic tool for early detection, metastatic assessment and therapeutic monitoring of HER-2 positive cancers.


Subject(s)
Antibodies/pharmacology , Breast Neoplasms/diagnostic imaging , Nanoparticles/chemistry , Receptor, ErbB-2/immunology , A549 Cells , Animals , Breast Neoplasms/pathology , Drug Delivery Systems , Gadolinium , Humans , Kinetics , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Tomography, X-Ray Computed/methods
6.
Vaccine ; 38(19): 3537-3544, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32245642

ABSTRACT

BACKGROUND: Vaccination is the most effective approach to prevent infection with highly pathogenic avian influenza (HPAI). Adjuvants are often used to induce effective immune responses and overcome the immunological weakness of recombinant HPAI antigens. Given the logistical challenges of immunization to HPAI during pandemic situations, vaccines administered via the intramuscular (I.M.) route would be of value. METHODS: A new formulation of nanoemulsion adjuvant (NE02) suitable for I.M. vaccination was developed. This NE02 was evaluated alone and in combination with CpG to develop H5 immune responses in mouse and ferret models. Measures of recombinant H5 (rH5) specific immunity evaluated included serum IgG and IgG subclasses, bronchoalveolar lavage fluid IgA, and cytokines. The activation of NF-kB was also analyzed. The efficacy of the vaccine was assessed by performing hemagglutination inhibition (HAI), virus neutralization (VN) assays, and viral challenges in ferrets. RESULTS: I.M. vaccination with rH5-NE02 significantly increased rH5-specific IgG and protected ferrets in the viral challenge model providing complete protection and sterile immunity in all animals tested. Combining NE02 and CpG produced accelerated antibody responses and this was accompanied by an elevation of IFN-γ and IL-17 responses and the downregulation of IL-5. The combination also caused a synergistic effect on NF-kB activation. In immunized ferrets after viral challenge, the rH5-NE02 + CpG vaccine via I.M. achieved at least 75% and 88% seroconversion of HAI and VN antibody responses, respectively, and improved body temperature stabilization and weight loss over NE02 alone. CONCLUSIONS: The I.M. injection of NE02 adjuvanted rH5 elicits strong and broad immune responses against H5 antigens and effectively protects animals from lethal H5 challenge. Combining this adjuvant with CpG enhanced immune responses and provided improvements in outcomes to viral challenge in ferrets. The results suggest that combinations of adjuvants may be useful to enhance H5 immune responses and improve protection against influenza infection.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza Vaccines , Orthomyxoviridae Infections , Adjuvants, Immunologic , Animals , Antibodies, Viral , Ferrets , Mice , Orthomyxoviridae Infections/prevention & control
7.
mSphere ; 4(6)2019 12 18.
Article in English | MEDLINE | ID: mdl-31852807

ABSTRACT

Infections triggered by pathogenic fungi cause a serious threat to the public health care system. In particular, an increase of antifungal drug-resistant fungi has resulted in difficulty in treatment. A limited variety of antifungal drugs available to treat patients has left us in a situation where we need to develop new therapeutic approaches that are less prone to development of resistance by pathogenic fungi. In this study, we demonstrate the efficacy of the nanoemulsion NB-201, which utilizes the surfactant benzalkonium chloride, against human-pathogenic fungi. We found that NB-201 exhibited in vitro activity against Candidaalbicans, including both planktonic growth and biofilms. Furthermore, treatments with NB-201 significantly reduced the fungal burden at the infection site and presented an enhanced healing process after subcutaneous infections by multidrug-resistant C. albicans in a murine host system. NB-201 also exhibited in vitro growth inhibition activity against other fungal pathogens, including Cryptococcus spp., Aspergillus fumigatus, and Mucorales Due to the nature of the activity of this nanoemulsion, there is a minimized chance of drug resistance developing, presenting a novel treatment to control fungal wound or skin infections.IMPORTANCE Advances in medicine have resulted in the discovery and implementation of treatments for human disease. While these recent advances have been beneficial, procedures such as solid-organ transplants and cancer treatments have left many patients in an immunocompromised state. Furthermore, the emergence of immunocompromising diseases such as HIV/AIDS or other immunosuppressive medical conditions have opened an opportunity for fungal infections to afflict patients globally. The development of drug resistance in human-pathogenic fungi and the limited array of antifungal drugs has left us in a scenario where we need to develop new therapeutic approaches to treat fungal infections that are less prone to the development of resistance by pathogenic fungi. The significance of our work lies in utilizing a novel nanoemulsion formulation to treat topical fungal infections while minimizing risks of drug resistance development.


Subject(s)
Antifungal Agents/pharmacology , Benzalkonium Compounds/pharmacology , Fungi/drug effects , Polysorbates/pharmacology , Soybean Oil/pharmacology , Animals , Antifungal Agents/administration & dosage , Benzalkonium Compounds/administration & dosage , Candidiasis/drug therapy , Disease Models, Animal , Drug Combinations , Mice , Microbial Sensitivity Tests , Polysorbates/administration & dosage , Soybean Oil/administration & dosage , Treatment Outcome
8.
Vaccine ; 37(12): 1591-1600, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30795941

ABSTRACT

BACKGROUND: Highly pathogenic H5N1 influenza viruses remain a pandemic risk to the world population. Although vaccines are the best solution to prevent this threat, a more effective vaccine for H5 strains of influenza has yet to be developed. All existing vaccines target only serum antibody against influenza as the primary outcome, while mucosal immunity has not been addressed. To address these shortcomings we have used an effective mucosal adjuvant system to produce a prototype vaccine that provides antibody, cellular and mucosal immunity to multiple serotypes of H5. METHODS: Plant-derived recombinant H5 (rH5) antigen was mixed with a novel nanoemulsion NE01 adjuvant. The rH5-NE01 vaccine was administered intranasally to CD-1 mice and ferrets. Immunogenicity of this immunization was evaluated through rH5-specific antibody and cellular immune responses. Hemagglutination inhibition (HI) and virus neutralization (VN) assays were performed. Protection against H5N1 virus challenge was evaluated in ferrets. RESULTS: Intranasal immunization with rH5-NE01vaccine induced high titers (>106) of rH5-specific IgG in mice. In mice and ferrets this vaccine also achieved titers of ≥40 for both HI and VN. Additionally, the levels of rH5-specific IgA were significantly increased in bronchial secretions in these animals. The rH5-NE01 vaccine enhanced rH5-specific cellular immune responses including IFN-γ and IL-17. Ten-day survival post challenge was 100% in ferrets that received rH5-NE01compared to 12.5% in the PBS group. Furthermore, this vaccine prevented weight loss and increases in body temperature after H5N1 challenge as compared to the controls. Moreover, H5N1 virus in nasal wash of rH5-NE01-vaccinated ferrets was significantly decreased compared to controls. CONCLUSION: Intranasal immunization with rH5 antigen formulated with NE01 adjuvant elicited strong, broad and balanced immune responses that effectively protect against H5N1 influenza virus infection in the ferret model. The ease of formulation of rH5-NE01 makes this novel combination a promising mucosal vaccine candidate for pandemic influenza.


Subject(s)
Adjuvants, Immunologic , Emulsions , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cytokines/metabolism , Female , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Immunization , Immunogenicity, Vaccine , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Male , Mice , Orthomyxoviridae Infections/prevention & control , Recombinant Proteins
9.
Nanomedicine (Lond) ; 12(10): 1177-1185, 2017 May.
Article in English | MEDLINE | ID: mdl-28447896

ABSTRACT

AIM: To develop NB-201, a nanoemulsion compound, as a novel microbicidal agent against methicillin-resistant Staphylococcus aureus (MRSA) infection, which is a common threat to public health but with limited therapeutic options. MATERIALS & METHODS: NB-201 was tested in in vitro and in vivo murine and porcine models infected with MRSA. RESULTS: Topical treatment of MRSA-infected wounds with NB-201 significantly decreased bacterial load and had no toxic effects on healthy skin tissues. NB-201 attenuated neutrophil sequestration in MRSA-infected wounds and inhibited epidermal and deep dermal inflammation. The levels of proinflammatory cytokines were reduced in NB-201-treated MRSA-infected wounds. CONCLUSION: NB-201 can greatly reduce inflammation characteristic of infected wounds and has antimicrobial activity that effectively kills MRSA regardless of the genetic basis of antibiotic resistance.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Benzalkonium Compounds/therapeutic use , Methicillin-Resistant Staphylococcus aureus/drug effects , Polysorbates/therapeutic use , Soybean Oil/therapeutic use , Staphylococcal Infections/drug therapy , Wound Infection/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Benzalkonium Compounds/pharmacology , Cytokines/analysis , Drug Combinations , Female , Humans , Mice , Microbial Sensitivity Tests , Polysorbates/pharmacology , Soybean Oil/pharmacology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Swine , Wound Infection/microbiology , Wound Infection/pathology
10.
Mil Med ; 181(5 Suppl): 259-64, 2016 05.
Article in English | MEDLINE | ID: mdl-27168582

ABSTRACT

Despite advances in antimicrobial therapies, wound infection remains a global public health concern. We aimed to formulate and assess various nanoemulsions (NEs) for potential effectiveness as stable antimicrobial agents suitable for topic application. A total of 106 NEs were developed that varied with respect to nonionic and cationic surfactants. Stability testing demonstrated that the NEs tested are broadly stable, with 97/106 formulations passing 2-week stability tests. Two NEs, NB-201 and NB-402, were selected to test antimicrobial activity in a wound model in mice. Skin abrasion wounds were infected with Staphylococcus aureus followed by NE treatment. Infected skin was then evaluated by measuring colony forming units. NB-201 reduced median bacterial counts by 4 to 5 log compared to animals treated with saline, whereas NB-402 reduced bacterial counts by 2 to 3 log. Additional stability tests on NB-201 demonstrated that NB-201 is stable in the presence of human serum, and is stable for at least 6 months at 5°C, 25°C, and 40°C. Finally, in in vitro studies, NB-201 was found to be effective against S. aureus at a higher dilution than the commercially available silver sulfadiazine. Altogether these results demonstrate that NB-201 is a stable and effective topical antimicrobial for the treatment of S. aureus.


Subject(s)
Benzalkonium Compounds/pharmacology , Cetylpyridinium/pharmacology , Poloxamer/pharmacology , Polysorbates/pharmacology , Soybean Oil/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Animals , Anti-Infective Agents, Local/administration & dosage , Anti-Infective Agents, Local/pharmacology , Anti-Infective Agents, Local/therapeutic use , Benzalkonium Compounds/administration & dosage , Benzalkonium Compounds/therapeutic use , Cetylpyridinium/administration & dosage , Cetylpyridinium/therapeutic use , Drug Combinations , Mice , Models, Animal , Poloxamer/administration & dosage , Poloxamer/therapeutic use , Polysorbates/administration & dosage , Polysorbates/therapeutic use , Silver Sulfadiazine/administration & dosage , Silver Sulfadiazine/pharmacology , Silver Sulfadiazine/therapeutic use , Soybean Oil/administration & dosage , Soybean Oil/therapeutic use , Wound Infection/drug therapy , Wound Infection/prevention & control
11.
Oncotarget ; 7(24): 36002-36013, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27144519

ABSTRACT

Although many breast and lung cancers overexpress human epidermal growth factor receptor-2 (HER-2), no methods currently exist for effective and early detection of HER-2-positive cancers. To address this issue, we designed and synthesized dendrimer-based novel nano-imaging agents that contain gold nanoparticles (AuNPs) and gadolinium (Gd), conjugated with the humanized anti-HER-2 antibody (Herceptin). Generation 5 (G5) polyamidoamine (PAMAM) dendrimers were selected as the backbone for the nano-imaging agents due to their unique size, high ratio of surface functional groups and bio-functionality. We modified G5 PAMAM dendrimer surface with PEG and 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelators to encapsulate AuNPs and complex Gd. These dendrimer entrapped AuNPs were further conjugated with Herceptin through copper-catalyzed azide- alkyne click reaction to construct the nano-imaging agent Au-G5-Gd-Herceptin. The targeted nano-imaging agent bound selectively to HER-2 overexpressing cell lines, with subsequent internalization into the cells. More importantly, non-targeted nano-imaging agent neither bound nor internalized into cells overexpressing HER-2. These results suggest that our approach could provide a platform to develop nano-diagnostic agents or nano-therapeutic agents for early detection and treatment of HER-2-positive cancers.


Subject(s)
Cell Tracking/methods , Dendrimers/chemistry , Metal Nanoparticles/chemistry , Receptor, ErbB-2/metabolism , Trastuzumab/metabolism , A549 Cells , Cell Line, Tumor , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Microscopy, Confocal , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Trastuzumab/chemistry , Trastuzumab/immunology
12.
Nanomaterials (Basel) ; 6(12)2016 Nov 25.
Article in English | MEDLINE | ID: mdl-28335351

ABSTRACT

A nanoemulsion (NE) is a surfactant-based, oil-in-water, nanoscale, high-energy emulsion with a mean droplet diameter of 400-600 nm. When mixed with antigen and applied nasally, a NE acts as a mucosal adjuvant and induces mucosal immune responses. One possible mechanism for the adjuvant effect of this material is that it augments antigen uptake and distribution to lymphoid tissues, where the immune response is generated. Biocompatible iron oxide nanoparticles have been used as a unique imaging approach to study the dynamics of cells or molecular migration. To study the uptake of NEs and track them in vivo, iron oxide nanoparticles were synthesized and dispersed in soybean oil to make iron oxide-modified NEs. Our results show that iron oxide nanoparticles can be stabilized in the oil phase of the nanoemulsion at a concentration of 30 µg/µL and the iron oxide-modified NEs have a mean diameter of 521 nm. In vitro experiments demonstrated that iron oxide-modified NEs can affect uptake by TC-1 cells (a murine epithelial cell line) and reduce the intensity of magnetic resonance (MR) images by shortening the T2 time. Most importantly, in vivo studies demonstrated that iron oxide-modified NE could be detected in mouse nasal septum by both transmission electron microscopy and MR imaging. Altogether these experiments demonstrate that iron oxide-modified NE is a unique tool that can be used to study uptake and distribution of NEs after nasal application.

13.
J Transl Med ; 12: 180, 2014 Jun 23.
Article in English | MEDLINE | ID: mdl-24957380

ABSTRACT

BACKGROUND: BID functions as a bridge molecule between death-receptor and mitochondrial related apoptotic pathways to amplify apoptotic signaling. Our previous studies have demonstrated a substantial increase in BID expression in primary normal thyroid epithelia cells treated with inflammatory cytokines, including the combination of IFNγ and IL-1ß or IFNγ and TNFα. The aim of this study was to determine whether an increase in BID expression in thyroid can induce autoimmune thyroiditis. METHODS: A transgenic mouse line that expresses human BID in thyroid cells was established by fusing a mouse thyroglobulin (Tg) promoter upstream of human BID (Tg-BID). We tested whether the increased expression of pro-apoptotic BID in thyroid would induce autoimmune thyroiditis, both in the presence and absence of 0.3% iodine water. RESULTS: Our data show that Tg-BID mice in a CBA/J (H-2 k) background do not spontaneously develop autoimmune thyroiditis for over a year. However, upon ingestion of iodine in the drinking water, autoimmune thyroiditis does develop in Tg-BID transgenic mice, as shown by a significant increase in anti-Tg antibody and mononuclear cell infiltration in the thyroid glands in 30% of mice tested. Serum T4 levels, however, were similar between iodine-treated Tg-BID transgenic mice and the wild type mice. CONCLUSIONS: Our data demonstrate that increased thyroid expression of BID facilitates the development of autoimmune thyroiditis induced by iodine uptake. However, the overexpression of BID itself is not sufficient to initiate thyroiditis in CBA/J (H-2 k) mice.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Iodine/adverse effects , Thyroid Gland/metabolism , Thyroiditis, Autoimmune/metabolism , Animals , BH3 Interacting Domain Death Agonist Protein/genetics , Base Sequence , DNA Primers , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Thyroiditis, Autoimmune/chemically induced
14.
Mol Pharm ; 11(2): 531-44, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24320221

ABSTRACT

The development of effective intranasal vaccines is of great interest due to their potential to induce both mucosal and systemic immunity. Here we produced oil-in-water nanoemulsion (NE) formulations containing various cationic and nonionic surfactants for use as adjuvants for the intranasal delivery of vaccine antigens. NE induced immunogenicity and antigen delivery are believed to be facilitated through initial contact interactions between the NE droplet and mucosal surfaces which promote prolonged residence of the vaccine at the site of application, and thus cellular uptake. However, the details of this mechanism have yet to be fully characterized experimentally. We have studied the physicochemical properties of the NE droplet surfactant components and demonstrate that properties such as charge and polar headgroup geometry influence the association of the adjuvant with the mucus protein, mucin. Association of NE droplets with mucin in vitro was characterized by various biophysical and imaging methods including dynamic light scattering (DLS), zeta potential (ZP), and surface plasmon resonance (SPR) measurements as well as transmission electron microscopy (TEM). Emulsion surfactant compositions were varied in a systematic manner to evaluate the effects of hydrophobicity and polar group charge/size on the NE-mucin interaction. Several cationic NE formulations were found to facilitate cellular uptake of the model antigen, ovalbumin (OVA), in a nasal epithelial cell line. Furthermore, fluorescent images of tissue sections from mice intranasally immunized with the same NEs containing green fluorescent protein (GFP) antigen demonstrated that these NEs also enhanced mucosal layer penetration and cellular uptake of antigen in vivo. NE-mucin interactions observed through biophysical measurements corresponded with the ability of the NE to enhance cellular uptake. Formulations that enhanced antigen uptake in vitro and in vivo also led to the induction of a more consistent antigen specific immune response in mice immunized with NEs containing OVA, linking NE-facilitated mucosal layer penetration and cellular uptake to enhancement of the immune response. These findings suggest that biophysical measurement of the mucoadhesive properties of emulsion based vaccines constitutes an effective in vitro strategy for selecting NE candidates for further evaluation in vivo as mucosal adjuvants.


Subject(s)
Adjuvants, Immunologic/chemistry , Emulsions/chemistry , Emulsions/pharmacology , Nasal Mucosa/drug effects , Surface-Active Agents/chemistry , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Cell Adhesion/drug effects , Cell Line , Chemistry, Pharmaceutical , Female , Humans , Immunogenetic Phenomena , Mice , Microscopy, Electron, Transmission , Nanotechnology , Surface-Active Agents/pharmacology
15.
Int J Biol Sci ; 10(1): 25-32, 2013.
Article in English | MEDLINE | ID: mdl-24391448

ABSTRACT

HER2 is an oncogenic tumor-associated antigen overexpressed in 20-25% of breast cancers, which is associated with increased invasion, metastasis of the disease and resistance to therapy. Recent studies have further shown that HER2 can increase the population of breast cancer stem cells (BCSCs). However, there is currently no in vivo model for the study of HER2(+) BCSCs. In this study, we characterized a mouse breast cancer model for HER2(+) BCSCs. This was accomplished by inoculating mouse mammary tumor EO771 cells engineered with human wild-type HER2 (EO771E2) into C57BL/6 HER2 transgenic mice to test and confirm the stable human HER2 expression in the model. More importantly, we detected a subpopulation of EO771E2 cells with a high activity of aldehyde dehydrogenases (ALDH(high)). We demonstrated that the isolated ALDH(high) EO771E2 cells possessed key properties of BCSCs including enhanced tumorigenicity, generation of heterogeneous tumors and the capacity to self-renewal in vitro. In conclusion, the tumors formed in C57BL/6 HER2 transgenic mice with EO771E2 cell injection revealed stable and functional human HER2 expression. These tumors contain a subset of ALDH(high) cells which are small in number, but are enriched in cancer stem cells. This model is deemed to be useful for experiments aimed to develop novel treatments to target HER2(+) BCSCs.


Subject(s)
Breast Neoplasms/pathology , Genes, erbB-2 , Neoplastic Stem Cells/metabolism , Animals , Base Sequence , Breast Neoplasms/metabolism , DNA Primers , Disease Models, Animal , Female , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polymerase Chain Reaction
16.
Nanomedicine (Lond) ; 7(6): 867-76, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22420425

ABSTRACT

AIM: The aim of this study was to investigate the impact of a novel nanoemulsion (NE) adjuvant, a soybean oil emulsion, on autoimmune response. To this end, we used murine thyroglobulin (mTg)-induced experimental autoimmune thyroiditis in mice as a study model. MATERIALS & METHODS: Mice received NE or NE + mTg by nasal delivery. At 1 week after the second nasal delivery of NE with or without mTg, all mice were immunized with mTg and lipopolysaccharides to induce experimental autoimmune thyroiditis. RESULTS: Compared with controls, mTg-NE-treated mice had much more antigens accumulated in the nasal passage and thymus and developed a milder form of thyroiditis. This was accompanied by an increase in IL-10, IL-17 and reduced IFN-γ. The production of anti-mTg antibodies was significantly decreased in mTg-NE-treated mice. The percentage of Tregs in cervical lymph nodes was higher in mTg-NE-treated mice than NE-treated mice. Furthermore, Foxp3 and TGF-ß levels were prominently enhanced in mTg-NE-treated mice. CONCLUSION: This study indicates that a low dose of mTg in NE can significantly enhance antigen uptake and Tregs, resulting in inhibition of experimental autoimmune thyroiditis development.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Emulsions/therapeutic use , Immune Tolerance , Soybean Oil/therapeutic use , Thyroglobulin/administration & dosage , Thyroglobulin/immunology , Thyroiditis, Autoimmune/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Autoantibodies/immunology , Emulsions/administration & dosage , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-17/immunology , Mice , Mice, Inbred CBA , RNA, Messenger/genetics , Soybean Oil/administration & dosage , Soybean Oil/immunology , T-Lymphocytes, Regulatory/immunology , Thyroid Gland/immunology , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyroiditis, Autoimmune/immunology , Thyroiditis, Autoimmune/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
17.
Nanotechnology ; 23(10): 105601, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22349004

ABSTRACT

We report a facile approach to synthesizing 3-aminopropyltrimethoxysilane (APTS)-coated magnetic iron oxide (Fe(3)O(4)@APTS) nanoparticles (NPs) with tunable surface functional groups for potential biomedical applications. The Fe(3)O(4) NPs with a mean diameter of 6.5 nm were synthesized by a hydrothermal route in the presence of APTS. The formed amine-surfaced Fe(3)O(4)@APTS NPs were further chemically modified with acetic anhydride and succinic anhydride to generate neutral (Fe(3)O(4)@APTS⋅Ac) and negatively charged (Fe(3)O(4)@APTS⋅SAH) NPs. These differently functionalized NPs were extensively characterized by x-ray diffraction, transmission electron microscopy, Fourier transform infrared spectroscopy, thermogravimetry analysis, zeta potential measurements, and T(2) relaxometry. The cytotoxicity of the particles was evaluated by in vitro 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide colorimetric viability assay of cells along with microscopic observation of cell morphology. The hemocompatibility of the particles was assessed by in vitro hemolysis assay. We show that the hydrothermal approach enables an efficient modification of APTS onto the Fe(3)O(4) NP surfaces and the formed NPs with different surface charge polarities are water-dispersible and colloidally stable. The acetylated Fe(3)O(4)@APTS⋅Ac NPs displayed good biocompatibility and hemocompatibility in the concentration range of 0-100 µg ml(-1), while the pristine Fe(3)O(4)@APTS and Fe(3)O(4)@APTS⋅SAH particles started to display slight cytotoxicity at a concentration of 10 µg ml(-1). The findings from this study suggest that the Fe(3)O(4)@APTS NPs synthesized by the one-pot hydrothermal route can be surface modified for various potential biomedical applications.


Subject(s)
Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Nanotechnology/methods , Propylamines/chemistry , Propylamines/toxicity , Silanes/chemistry , Silanes/toxicity , Acetic Anhydrides , Cell Survival/drug effects , Erythrocytes , Hemolysis/drug effects , Humans , KB Cells , Magnetic Resonance Spectroscopy , Particle Size , Succinic Anhydrides , Surface Properties
18.
Soft Matter ; 6(11): 2539-2545, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20852741

ABSTRACT

We report the complexation of a potential anticancer agent 2-methoxyestradiol (2-ME) with generation 5 (G5) poly(amidoamine) dendrimers having different surface functional groups for therapeutic applications. The complexation experiment shows that approximately 6-8 drug molecules can be complexed with one dendrimer molecule regardless the type of the dendrimer terminal groups. The bioactivity of 2-ME complexed with dendrimers was found to be significantly dependent on the surface charge of G5 dendrimers. In vitro cell biological assays show that amine, hydroxyl, and acetamide-terminated G5 dendrimers with positive, slightly positive, and close to neutral surface charges, respectively are able to deliver 2-ME to inhibit cancer cell growth. In contrast, 2-ME complexed with carboxyl-terminated G5 dendrimers with negative surface charges does not show its inherent bioactivity. Further molecular dynamics simulation studies show that the compact structure of carboxylated G5 dendrimers complexed with 2-ME does not allow the release of the drug molecules even at a pH of 5.0, which is the typical pH value in lysosome. Our findings indicate that the surface modification of dendrimers with different charges is crucial for the development of formulations of various anticancer drugs for therapeutic applications.

19.
Biopolymers ; 91(11): 936-42, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19603491

ABSTRACT

Dendrimer-based nanotechnology significantly advances the area of targeted cancer imaging and therapy. Herein, we compared the difference of surface acetylated fluorescein isocyanate (FI) and folic acid (FA) modified generation 5 (G5) poly(amidoamine) dendrimers (G5.NHAc-FI-FA), and dendrimer-entrapped gold nanoparticles with similar modifications ([(Au(0))(51.2)-G5.NHAc-FI-FA]) in terms of their specific internalization to FA receptor (FAR)-overexpressing cancer cells. Confocal microscopic studies show that both G5.NHAc-FI-FA and [(Au(0))(51.2-)G5.NHAc-FI-FA] exhibit similar internalization kinetics regardless of the existence of Au nanoparticles (NPs). Molecular dynamics simulation of the two different nanostructures reveals that the surface area and the FA moiety distribution from the center of the geometry are slightly different. This slight difference may not be recognized by the FARs on the cell membrane, consequently leading to similar internalization kinetics. This study underlines the fact that metal or inorganic NPs entrapped within dendrimers interact with cells in a similar way to that of dendrimers lacking host NPs.


Subject(s)
Biocompatible Materials/pharmacokinetics , Carrier Proteins/metabolism , Dendrimers/pharmacokinetics , Endocytosis , Folic Acid/metabolism , Gold/pharmacokinetics , Metal Nanoparticles , Receptors, Cell Surface/metabolism , Antineoplastic Agents/administration & dosage , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Dendrimers/chemical synthesis , Dendrimers/chemistry , Drug Delivery Systems/methods , Ethylenediamines/chemistry , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Folate Receptors, GPI-Anchored , Folic Acid/chemistry , Gold/chemistry , Humans , KB Cells , Kinetics , Metal Nanoparticles/chemistry , Models, Molecular , Molecular Dynamics Simulation , Polyamines/chemistry , Thiourea/analogs & derivatives , Thiourea/chemistry
20.
Analyst ; 134(7): 1373-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19562204

ABSTRACT

We develop a facile approach to fabricating multifunctional dendrimer-stabilized gold nanoparticles (Au DSNPs) for cancer cell targeting and imaging. In this work, amine-terminated generation 5 (G5) poly(amidoamine) (PAMAM) dendrimers pre-functionalized with folic acid (FA) and fluorescein isothiocyanate (FI) are complexed with Au(III) ions, followed by acetylation of the amine groups on the dendrimer surfaces. This one-step process leads to the spontaneous formation of 6 nm-sized Au nanoparticles stabilized by multifunctional dendrimers bearing both targeting and imaging functionalities. The multifunctional Au DSNPs are characterized by UV-Vis spectrometry, 1H NMR, and transmission electron microscopy (TEM). The formed Au DSNPs are water-soluble, stable, and biocompatible. Combined flow cytometry, confocal microscopy, silver staining, and inductively coupled plasma-mass spectrometry (ICP-MS) analyses show that the FA- and FI-functionalized Au DSNPs can specifically target to cancer cells expressing high-affinity FA receptors in vitro. This approach to functionalizing Au DSNPs may be extended to other targeting molecules, providing a unique nanoplatform for targeting and imaging of a variety of biological systems.


Subject(s)
Dendrimers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Molecular Imaging/methods , Neoplasms/pathology , Cell Survival/drug effects , Gold/metabolism , Gold/toxicity , Humans , KB Cells , Neoplasms/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...