Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
2.
Biosensors (Basel) ; 5(4): 750-67, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26690235

ABSTRACT

The identification and characterization, at the cellular level, of cytokine productions present a high interest for both fundamental research and clinical studies. However, the majority of techniques currently available (ELISA, ELISpot, flow cytometry, etc.) have several shortcomings including, notably, the assessment of several cytokines in relation to individual secreting cells and the monitoring of living cell responses for a long incubation time. In the present work, we describe a system composed of a microfluidic platform coupled with an antibody microarray chip for continuous SPR imaging and immunofluorescence analysis of cytokines (IL-2 and IFN-γ) secreted by T-Lymphocytes, specifically, and stably captured on the biochip under flow upon continued long-term on-chip culture (more than 24 h).


Subject(s)
Antibodies, Immobilized/chemistry , Interferon-gamma/analysis , Interleukin-2/analysis , Lab-On-A-Chip Devices , Surface Plasmon Resonance/instrumentation , T-Lymphocytes/immunology , Adult , Antibodies, Immobilized/immunology , Cell Survival , Equipment Design , Humans , Interferon-gamma/immunology , Interleukin-2/immunology , Protein Array Analysis/instrumentation , T-Lymphocytes/chemistry
3.
Int J Nanomedicine ; 8: 2085-99, 2013.
Article in English | MEDLINE | ID: mdl-23785233

ABSTRACT

We recently demonstrated by in vitro experiments that PLGA (poly D, L-lactide-co-glycolide) potentiates T helper 1 (Th1) immune responses induced by a peptide derived from the recombinant major outer membrane protein (rMOMP) of Chlamydia trachomatis, and may be a promising vaccine delivery system. Herein we evaluated the immune-potentiating potential of PLGA by encapsulating the full-length rMOMP (PLGA-rMOMP), characterizing it in vitro, and investigating its immunogenicity in vivo. Our hypothesis was that PLGA-rMOMP triggers Th1 immune responses in mice, which are desirable prerequisites for a C. trachomatis candidate nanovaccine. Physical-structural characterizations of PLGA-rMOMP revealed its size (approximately 272 nm), zeta potential (-14.30 mV), apparent spherical smooth morphology, and continuous slow release pattern. PLGA potentiated the ability of encapsulated rMOMP to trigger production of cytokines and chemokines by mouse J774 macrophages. Flow cytometric analyses revealed that spleen cells from BALB/c mice immunized with PLGA-rMOMP had elevated numbers of CD4+ and CD8+ T cell subsets, and secreted more rMOMP-specific interferon-gamma (Th1) and interleukin (IL)-12p40 (Th1/Th17) than IL-4 and IL-10 (Th2) cytokines. PLGA-rMOMP-immunized mice produced higher serum immunoglobulin (Ig)G and IgG2a (Th1) than IgG1 (Th2) rMOMP-specific antibodies. Notably, sera from PLGA-rMOMP-immunized mice had a 64-fold higher Th1 than Th2 antibody titer, whereas mice immunized with rMOMP in Freund's adjuvant had only a four-fold higher Th1 than Th2 antibody titer, suggesting primarily induction of a Th1 antibody response in PLGA-rMOMP-immunized mice. Our data underscore PLGA as an effective delivery system for a C. trachomatis vaccine. The capacity of PLGA-rMOMP to trigger primarily Th1 immune responses in mice promotes it as a highly desirable candidate nanovaccine against C. trachomatis.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia trachomatis/immunology , Nanoparticles/chemistry , Porins/immunology , Vaccines, Subunit/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/chemistry , Bacterial Vaccines/pharmacokinetics , Cell Line , Chemokines/analysis , Chemokines/metabolism , Cytokines/analysis , Cytokines/metabolism , Female , Flow Cytometry , Lactic Acid/chemistry , Macrophages , Mice , Mice, Inbred BALB C , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Porins/chemistry , Porins/pharmacokinetics , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Th1 Cells , Vaccines, Subunit/chemistry , Vaccines, Subunit/pharmacokinetics
4.
Int J Nanomedicine ; 8: 1759-71, 2013.
Article in English | MEDLINE | ID: mdl-23690681

ABSTRACT

Chlamydia trachomatis is a bacterial sexually transmitted infection affecting millions of people worldwide. Previous vaccination attempts have employed the recombinant major outer membrane protein (MOMP) of C. trachomatis nonetheless, with limited success, perhaps, due to stability, degradation, and delivery issues. In this study we cloned C. trachomatis recombinant MOMP DNA (DMOMP) and encapsulated it in chitosan nanoparticles (DMCNP) using the complex coacervation technique. Physiochemical characterizations of DMCNP included transmission and scanning electron microcopy, Fourier transform infrared and ultraviolet-visible spectroscopy, and zeta potential. Encapsulated DMOMP was 167-250 nm, with a uniform spherical shape and homogenous morphology, and an encapsulation efficiency > 90%. A slow release pattern of encapsulated DMOMP, especially in acidic solution, was observed over 7 days. The zeta potential of DMCNP was ~8.80 mV, which indicated that it was highly stable. Toxicity studies of DMCNP (25-400 µg/mL) to Cos-7 cells using the MTT assay revealed minimal toxicity over 24-72 hours with >90% viable cells. Ultra-violet visible (UV-vis) spectra indicated encapsulated DMOMP protection by chitosan, whereas agarose gel electrophoresis verified its protection from enzymatic degradation. Expression of MOMP protein in DMCNP-transfected Cos-7 cells was demonstrated via Western blotting and immunofluorescence microscopy. Significantly, intramuscular injection of BALB/c mice with DMCNP confirmed the delivery of encapsulated DMOMP, and expression of the MOMP gene transcript in thigh muscles and spleens. Our data show that encapsulation of DMOMP in biodegradable chitosan nanoparticles imparts stability and protection from enzymatic digestion, and enhances delivery and expression of DMOMP in vitro and in mice. Further investigations of the nanoencapsulated DMCNP vaccine formulation against C. trachomatis in mice are warranted.


Subject(s)
Chitosan/chemistry , Chlamydia trachomatis/genetics , Drug Carriers/chemistry , Porins/genetics , Vaccines, DNA/chemistry , Animals , COS Cells , Cell Survival/drug effects , Chitosan/pharmacokinetics , Chitosan/pharmacology , Chlorocebus aethiops , DNA, Bacterial/genetics , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Stability , Female , Mice , Mice, Inbred BALB C , Nanoparticles , Particle Size , Porins/metabolism , RNA/analysis , RNA/isolation & purification , Vaccines, DNA/genetics , Vaccines, DNA/pharmacokinetics , Vaccines, DNA/pharmacology
5.
Mediators Inflamm ; 2012: 520174, 2012.
Article in English | MEDLINE | ID: mdl-22529524

ABSTRACT

Chlamydia trachomatis infects macrophages and epithelial cells evoking acute and chronic inflammatory conditions, which, if not controlled, may put patients at risk for major health issues such as pelvic inflammatory disease, chronic abdominal pain, and infertility. Here we hypothesized that IL-10, with anti-inflammatory properties, will inhibit inflammatory mediators that are produced by innate immune cells exposed to C. trachomatis. We used human epithelial (HeLa) cells and mouse J774 macrophages as target cells along with live and UV-inactivated C. trachomatis mouse pneumonitis (MoPn) as stimulants. Confocal microscopy employing an anti-Chlamydia antibody confirmed cells infectivity by day 1, which persisted up to day 3. Kinetics studies revealed that live C. trachomatis induced TNF, IL-6, and IL-8, as a function of time, with day-2 infection inducing the highest cytokine levels. Exogenous IL-10 inhibited TNF, IL-6, and IL-8 as secreted by day-2 infected cells. Similarly, IL-10 diminished cytokine levels as produced by macrophages exposed to UV-inactivated Chlamydia, suggesting the IL-10-mediated inhibition of cytokines is not restricted to live organisms. Our data imply that IL-10 is an important regulator of the initial inflammatory response to C. trachomatis infection and that further investigations be made into IL-10 use to combat inflammation induced by this bacterium.


Subject(s)
Chlamydia trachomatis/metabolism , Epithelial Cells/cytology , Interleukin-10/metabolism , Macrophages/cytology , Animals , Cytokines/metabolism , Dose-Response Relationship, Drug , Gene Expression Profiling , HeLa Cells , Humans , Inflammation , Interleukin-6/metabolism , Interleukin-8/metabolism , Mice , Microscopy, Confocal/methods , Recombinant Proteins/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...