Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 38717, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27958291

ABSTRACT

Interleukin (IL)-17 is one of the critical inflammatory cytokines that plays a direct role in development of Sjögren's syndrome (SjS), a systemic autoimmune disease characterized by a progressive chronic attack against the exocrine glands. The expression levels of IL-17 are correlated with a number of essential clinical parameters such as focus score and disease duration in human patients. Significantly immunological differences of Th17 cells were detected at the onset of clinical disease in female SjS mice compared to males. To further define the role of IL-17 in SjS and elucidate its involvement in the sexual dimorphism, we examined the systemic effect of IL-17 by genetically ablating Il-17 in the C57BL/6.NOD-Aec1Aec2, spontaneous SjS murine model. The results indicate that IL-17 is a potent inflammatory molecule in the induction of chemoattractants, cytokines, and glandular apoptosis in males and females. Elimination of IL-17 reduced sialadenitis more drastically in females than males. IL-17 is highly involved in modulating Th2 cytokines and altering autoantibody profiles which has a greater impact on changing plasma cells and germinal center B cell populations in females than males. The result supports a much more important role for IL-17 and demonstrates the sexual dimorphic function of IL-17 in SjS.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Interleukin-17/immunology , Salivary Glands/immunology , Sex Characteristics , Sjogren's Syndrome/immunology , Th2 Cells/immunology , Animals , B-Lymphocytes/pathology , Disease Models, Animal , Female , Germinal Center/pathology , Interleukin-17/genetics , Male , Mice , Salivary Glands/pathology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , Th2 Cells/pathology
2.
Curr Gene Ther ; 16(2): 83-9, 2016.
Article in English | MEDLINE | ID: mdl-27029944

ABSTRACT

Gene electrotransfer is an effective approach for delivering plasmid DNA to a variety of tissues. Delivery of molecules with electric pulses requires control of the electrical parameters to achieve effective delivery. Since discomfort or tissue damage may occur with high applied voltage, the reduction of the applied voltage while achieving the desired expression may be an important improvement. One possible approach is to combine electrotransfer with exogenously applied heat. Previous work performed in vitro demonstrated that increasing temperature before pulsing can enhance gene expression and made it possible to reduce electric fields while maintaining expression levels. In the study reported here, this combination was evaluated in vivo using a novel electrode device designed with an inserted laser for application of heat. The results obtained in this study demonstrated that increased temperature during electrotransfer increased expression or maintained expression with a reduction in applied voltage. With further optimization this approach may provide the basis for both a novel method and a novel instrument that may greatly enhance translation of gene electrotransfer.


Subject(s)
Gene Transfer Techniques , Animals , Electrodes , Female , Gene Expression , Guinea Pigs , Temperature
3.
Bioelectrochemistry ; 103: 120-3, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25193443

ABSTRACT

The presence of increased temperature for gene electrotransfer has largely been considered negative. Many reports have published on the lack of heat from electrotransfer conditions to demonstrate that their effects are from the electrical pulses and not from a rise in temperature. Our hypothesis was to use low levels of maintained heat from an exogenous source to aid in gene electrotransfer. The goal was to increase gene expression and/or reduce electric field. In our study we evaluated high and low electric field conditions from 90 V to 45 V which had been preheated to 40 °C, 43 °C, or 45 °C. Control groups of non-heated as well as DNA only were included for comparison in all experiments. Luciferase gene expression, viability, and percent cell distribution were measured. Our results indicated a 2-4 fold increase in gene expression that is temperature and field dependent. In addition levels of gene expression can be increased without significant decreases in cell death and in the case of high electric fields no additional cell death. Finally, in all conditions percent cell distribution was increased from the application of heat. From these results, we conclude that various methods may be employed depending on the end user's desired goals. Electric field can be reduced 20-30% while maintaining or slightly increasing gene expression and increasing viability or overall gene expression and percent cell distribution can be increased with low viability.


Subject(s)
Electrochemical Techniques/methods , Gene Transfer Techniques , Cell Line , Cell Survival , Electroporation/methods , Gene Expression , Keratinocytes , Luciferases/genetics , Temperature
4.
Platelets ; 26(1): 13-6, 2015.
Article in English | MEDLINE | ID: mdl-24433255

ABSTRACT

Activation of human platelets produces a gel-like substance referred to as platelet rich plasma or platelet gel. Platelet gel is used clinically to promote wound healing; it also exhibits antimicrobial properties that may aid in the healing of infected wounds. The purpose of this study was to quantify the efficacy of human platelet gel against the opportunistic bacterial wound pathogens Acinetobacter baumannii, Pseudomonas aeruginosa, and Staphylococcus aureus on skin. These opportunistic pathogens may exhibit extensive antibiotic resistance, necessitating the development of alternative treatment options. The antimicrobial efficacy of platelet gel supernatants was quantified using an in vitro broth dilution assay, an ex vivo inoculated skin assay, and in an in vivo skin decontamination assay. Human platelet gel supernatants were highly bactericidal against A. baumannii and moderately but significantly bactericidal against S. aureus in vitro and in the ex vivo skin model. P. aeruginosa was not inactivated in vitro; a low but significant inactivation level was observed ex vivo. These supernatants were quite effective at inactivating a model organism on skin in vivo. These results suggest application of platelet gel has potential clinical applicability, not only in the acceleration of wound healing, but also against relevant bacteria causing wound infections.


Subject(s)
Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Blood Platelets/metabolism , Opportunistic Infections/drug therapy , Opportunistic Infections/microbiology , Platelet-Rich Plasma/metabolism , Wounds and Injuries/drug therapy , Wounds and Injuries/microbiology , Acinetobacter baumannii , Animals , Disease Models, Animal , Humans , Male , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Rats , Skin/injuries , Staphylococcus aureus , Wound Healing
5.
Expert Rev Clin Immunol ; 10(4): 469-81, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24506531

ABSTRACT

Sjögren's syndrome (SjS) is a complex chronic autoimmune disease of multifactorial etiology that results in eventual loss of secretory function in the exocrine glands. The challenges towards finding a therapeutic prevention or treatment for SjS are due primarily to a lack of understanding in the pathophysiological and clinical progression of the disease. In order to circumnavigate this problem, there is a need for appropriate animal models that resemble the major phenotypes of human SjS and deliver a clear underlying biological or molecular mechanism capable of defining various aspects for the disease. Here, we present an overview of SjS mouse models that are providing insight into the autoimmune process of SjS and advance our focus on potential diagnostic and therapeutic targets.


Subject(s)
Disease Models, Animal , Sjogren's Syndrome/immunology , Sjogren's Syndrome/pathology , Animals , Humans , Mice
6.
PLoS One ; 8(8): e73423, 2013.
Article in English | MEDLINE | ID: mdl-24015305

ABSTRACT

Topical gene delivery to the epidermis has the potential to be an effective therapy for skin disorders, cutaneous cancers, vaccinations and systemic metabolic diseases. Previously, we reported on a non-invasive multielectrode array (MEA) that efficiently delivered plasmid DNA and enhanced expression to the skin of several animal models by in vivo gene electrotransfer. Here, we characterized plasmid DNA delivery with the MEA in a hairless guinea pig model, which has a similar histology and structure to human skin. Significant elevation of gene expression up to 4 logs was achieved with intradermal DNA administration followed by topical non-invasive skin gene electrotransfer. This delivery produced gene expression in the skin of hairless guinea pig up to 12 to 15 days. Gene expression was observed exclusively in the epidermis. Skin gene electrotransfer with the MEA resulted in only minimal and mild skin changes. A low level of human Factor IX was detected in the plasma of hairless guinea pig after gene electrotransfer with the MEA, although a significant increase of Factor IX was obtained in the skin of animals. These results suggest gene electrotransfer with the MEA can be a safe, efficient, non-invasive skin delivery method for skin disorders, vaccinations and potential systemic diseases where low levels of gene products are sufficient.


Subject(s)
Electroporation/methods , Epidermis/metabolism , Gene Transfer Techniques , Plasmids/pharmacology , Skin Neoplasms/therapy , Animals , Epidermis/pathology , Factor IX/biosynthesis , Factor IX/genetics , Female , Guinea Pigs , Humans , Skin Neoplasms/genetics , Time Factors
7.
Bioelectrochemistry ; 94: 1-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23727769

ABSTRACT

Gene electrotransfer (GET) enhances delivery of DNA vaccines by increasing both gene expression and immune responses. Our lab has developed the multi-electrode array (MEA) for DNA delivery to skin. The MEA was used at constant pulse duration (150 ms) and frequency (6.67 Hz). In this study, delivery parameters including applied voltage (5-45 V), amount of plasmid (100-300 µg), and number of treatments (2-3) were evaluated for delivery of a DNA vaccine. Mice were intradermally injected with plasmid expressing Bacillus anthracis protective antigen with or without GET and αPA serum titers measured. Within this experiment no significant differences were noted in antibody levels from varying dose or treatment number. However, significant differences were measured from applied voltages of 25 and 35 V. These voltages generated antibody levels between 20,000 and 25,000. Serum from animals vaccinated with these conditions also resulted in toxin neutralization in 40-60% of animals. Visual damage was noted at MEA conditions of 40 V. No damage was noted either visually or histologically from conditions of 35 V or below. These results reflect the importance of establishing appropriate electrical parameters and the potential for the MEA in non-invasive DNA vaccination against B. anthracis.


Subject(s)
Anthrax/genetics , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Vaccines, DNA/immunology , Animals , Anthrax/prevention & control , Antigens, Bacterial/genetics , Antigens, Bacterial/toxicity , Bacillus anthracis/pathogenicity , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Electrodes , Gene Transfer Techniques , Humans , Mice , Vaccines, DNA/genetics
8.
PLoS One ; 6(4): e19181, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559474

ABSTRACT

Current progress in the development of vaccines has decreased the incidence of fatal and non-fatal infections and increased longevity. However, new technologies need to be developed to combat an emerging generation of infectious diseases. DNA vaccination has been demonstrated to have great potential for use with a wide variety of diseases. Alone, this technology does not generate a significant immune response for vaccination, but combined with delivery by electroporation (EP), can enhance plasmid expression and immunity. Most EP systems, while effective, can be invasive and painful making them less desirable for use in vaccination. Our lab recently developed a non-invasive electrode known as the multi-electrode array (MEA), which lies flat on the surface of the skin without penetrating the tissue. In this study we evaluated the MEA for its use in DNA vaccination using Hepatitis B virus as the infectious model. We utilized the guinea pig model because their skin is similar in thickness and morphology to humans. The plasmid encoding Hepatitis B surface antigen (HBsAg) was delivered intradermally with the MEA to guinea pig skin. The results show increased protein expression resulting from plasmid delivery using the MEA as compared to injection alone. Within 48 hours of treatment, there was an influx of cellular infiltrate in experimental groups. Humoral responses were also increased significantly in both duration and intensity as compared to injection only groups. While this electrode requires further study, our results suggest that the MEA has potential for use in electrically mediated intradermal DNA vaccination.


Subject(s)
Injections, Intradermal/methods , Vaccines, DNA/metabolism , Animals , Electrodes , Enzyme-Linked Immunosorbent Assay/methods , Equipment Design , Female , Guinea Pigs , Hepatitis B Surface Antigens/chemistry , Hepatitis B virus/metabolism , Humans , Immune System , Immunohistochemistry/methods , Models, Animal , Plasmids/metabolism , Skin/pathology , Vaccination
9.
J Control Release ; 151(3): 256-62, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21262290

ABSTRACT

Because of its large surface area and easy access for both delivery and monitoring, the skin is an attractive target for gene therapy for cutaneous diseases, vaccinations and several metabolic disorders. The critical factors for DNA delivery to the skin by electroporation (EP) are effective expression levels and minimal or no tissue damage. Here, we evaluated the non-invasive multielectrode array (MEA) for gene electrotransfer. For these studies we utilized a guinea pig model, which has been shown to have a similar thickness and structure to human skin. Our results demonstrate significantly increased gene expression 2 to 3 logs above injection of plasmid DNA alone over 15 days. Furthermore, gene expression could be enhanced by increasing the size of the treatment area. Transgene-expressing cells were observed exclusively in the epidermal layer of the skin. In contrast to caliper or plate electrodes, skin EP with the MEA greatly reduced muscle twitching and resulted in minimal and completely recoverable skin damage. These results suggest that EP with MEA can be an efficient and non-invasive skin delivery method with less adverse side effects than other EP delivery systems and promising clinical applications.


Subject(s)
DNA/administration & dosage , Electrochemotherapy/methods , Gene Transfer Techniques , Skin/metabolism , Animals , DNA/genetics , Electrochemotherapy/instrumentation , Electrodes , Electroporation , Female , Gene Expression , Green Fluorescent Proteins/genetics , Guinea Pigs , Injections, Intradermal , Luciferases/genetics , Plasmids/administration & dosage , Plasmids/genetics , Skin/pathology
10.
Article in English | MEDLINE | ID: mdl-22254415

ABSTRACT

A critical aspect of gene transfer is effective delivery of the transgene to the appropriate target. Electrically mediated delivery (electroporation) of plasmid DNA has been accepted as a viable approach to achieve effective delivery. One promising area is delivering plasmid DNA to skin. Gene transfer to the skin with electroporation is currently being evaluated for its potential for inducing angiogenesis for wound healing and for delivering DNA vaccines to the skin. Experiments utilizing a plasmid encoding for vascular endothelial growth factor has demonstrated how wound healing could be accelerated. In another study, delivery of a plasmid encoding Hepatitis B surface antigen have demonstrated that high antibody titers can be induced after two applications (prime/boost). Our laboratory has also examined the use of electroporation to delivery plasmid DNA encoding various cytokines as a potential therapy for melanoma. The plasmid is injected directly into the tumor followed by the administration of electroporation. Extensive preclinical work provided the rationale for a Phase I proof of concept first in human trial in patients with accessible cutaneous melanoma metastases. Biopsies of treated lesions showed significant necrosis of melanoma cells within the tumor as well as IL-12 expression. Lymphocytic infiltrate was observed in biopsies from patients in several cohorts. Clinical evidence of responses in untreated lesions suggested there was a systemic response following therapy was observed. Since this trial several other clinical studies utilizing electroporation to deliver plasmid DNA have been initiated. It is clear that this delivery approach has tremendous potential to facilitate the translation of gene transfer protocols from the bench to the bedside.


Subject(s)
DNA/administration & dosage , DNA/genetics , Electroporation/trends , Genetic Therapy/trends , Transfection/trends , Translational Research, Biomedical/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...