Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Radiol Case Rep ; 19(3): 1035-1039, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38226045

ABSTRACT

The authors report a case of pathologically proven intracardiac bronchogenic cyst embedded within the interatrial septum of a 30-year-old woman presenting with chest pain and first-degree AV block. Multimodality imaging played an essential role in the discovery, investigation, and diagnosis of this extremely rare entity.

3.
PLoS One ; 16(4): e0243683, 2021.
Article in English | MEDLINE | ID: mdl-33909614

ABSTRACT

Identification of genomic mutations by molecular testing plays an important role in diagnosis, prognosis, and treatment of myeloid neoplasms. Next-generation sequencing (NGS) is an efficient method for simultaneous detection of clinically significant genomic mutations with high sensitivity. Various NGS based in-house developed and commercial myeloid neoplasm panels have been integrated into routine clinical practice. However, some genes frequently mutated in myeloid malignancies are particularly difficult to sequence with NGS panels (e.g., CEBPA, CARL, and FLT3). We report development and validation of a 48-gene NGS panel that includes genes that are technically challenging for molecular profiling of myeloid neoplasms including acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN). Target regions were captured by hybridization with complementary biotinylated DNA baits, and NGS was performed on an Illumina NextSeq500 instrument. A bioinformatics pipeline that was developed in-house was used to detect single nucleotide variations (SNVs), insertions/deletions (indels), and FLT3 internal tandem duplications (FLT3-ITD). An analytical validation study was performed on 184 unique specimens for variants with allele frequencies ≥5%. Variants identified by the 48-gene panel were compared to those identified by a 35-gene hematologic neoplasms panel using an additional 137 unique specimens. The developed assay was applied to a large cohort (n = 2,053) of patients with suspected myeloid neoplasms. Analytical validation yielded 99.6% sensitivity (95% CI: 98.9-99.9%) and 100% specificity (95% CI: 100%). Concordance of variants detected by the 2 tested panels was 100%. Among patients with suspected myeloid neoplasms (n = 2,053), 54.5% patients harbored at least one clinically significant mutation: 77% in AML patients, 48% in MDS, and 45% in MPN. Together, these findings demonstrate that the assay can identify mutations associated with diagnosis, prognosis, and treatment options of myeloid neoplasms even in technically challenging genes.


Subject(s)
Hematologic Neoplasms , High-Throughput Nucleotide Sequencing , Leukemia, Myeloid, Acute , Mutation , Myelodysplastic Syndromes , Neoplasm Proteins , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
4.
J Diet Suppl ; 18(6): 597-613, 2021.
Article in English | MEDLINE | ID: mdl-33078636

ABSTRACT

INTRODUCTION: Supplementation with Rhodiola Rosea (RR) and Cordyceps Sinensis (CS) has been shown to improve aerobic performance, but their influence on concurrent training (resistance training plus high intensity interval training) outcomes has not been established. The purpose of this study was to determine the effects of supplementation with a multi-ingredient performance supplement (MIPS) containing RR and CS during a 14-week training and testing program on body composition, weekly exercise training outcomes, overall training and performance outcomes, and hormone profiles. METHODS: Active college-aged men (N = 21) were stratified into either a MIPS or a placebo (PLA) group. Both groups completed 14 weeks of training and testing. Body composition, overall training outcomes, and blood sample collection occurred at weeks 0, 7, and 14, while training performance was evaluated weekly. RESULTS: Both groups improved (p < 0.05) percent body fat (-1.3%), bench press (+4%) and squat strength (+8%), with no difference between groups. Serum cortisol concentrations significantly decreased (-11%) but there were no differences between groups. No other changes in blood hormone profiles occurred. Weekly exercise performance data suggests that MIPS improved sprint performance, bench press lifting volume, and total workload, but this did not lead to improved overall training performance compared to PLA over the14-week study. CONCLUSION: Despite MIPS improving certain aspects of weekly training performance, supplementation with MIPS for 14 weeks did not improve body composition, overall training and performance outcomes, or blood biomarkers of health in response to concurrent training in young men compared to PLA. This study was registered with clinicaltrials. gov (NCT02383017).


Subject(s)
Cordyceps , Resistance Training , Rhodiola , Body Composition , Dietary Supplements , Humans , Muscle Strength , Young Adult
5.
Vaccines (Basel) ; 8(4)2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33153189

ABSTRACT

Vaccines against blood-stage malaria often aim to induce antibodies to neutralize parasite entry into red blood cells, interferon gamma (IFNγ) produced by T helper 1 (Th1) CD4+ T cells or interleukin 4 (IL-4) produced by T helper 2 (Th2) cells to provide B cell help. One vaccine delivery method for suitable putative malaria protein antigens is the use of nanoparticles as vaccine carriers. It has been previously shown that antigen conjugated to inorganic nanoparticles in the viral-particle size range (~40-60 nm) can induce protective antibodies and T cells against malaria antigens in a rodent malaria challenge model. Herein, it is shown that biodegradable pullulan-coated iron oxide nanoparticles (pIONPs) can be synthesized in this same size range. The pIONPs are non-toxic and do not induce conventional pro-inflammatory cytokines in vitro and in vivo. We show that murine blood-stage antigen MSP4/5 from Plasmodium yoelii could be chemically conjugated to pIONPs and the use of these conjugates as immunogens led to the induction of both specific antibodies and IFNγ CD4+ T cells reactive to MSP4/5 in mice, comparable to responses to MSP4/5 mixed with classical adjuvants (e.g., CpG or Alum) that preferentially induce Th1 or Th2 cells individually. These results suggest that biodegradable pIONPs warrant further exploration as carriers for developing blood-stage malaria vaccines.

7.
Front Immunol ; 10: 331, 2019.
Article in English | MEDLINE | ID: mdl-30930890

ABSTRACT

Malaria remains a significant health problem in many tropical and sub-tropical regions. The development of vaccines against the clinically active blood-stage of infection needs to consider variability and polymorphism in target antigens, and an adjuvant system able to induce broad spectrum immunity comprising both antibodies and helper T cells. Moreover, recent studies have shown some conventional pro-inflammatory adjuvants can also promote expansion of immunosuppressive regulatory T cells (Treg) and myeloid derived suppressor cells (MDSC), both of which could negatively impact malaria disease progression. Herein, we explore the ability of a model nanoparticle delivery system (polystyrene nanoparticles; PSNPs), previously proven to not induce conventional inflammation, Treg or MDSC, to induce immunity to MSP4/5 from Plasmodium yoelii, a member of the MSP4 and MSP5 family of proteins which are highly conserved across diverse malaria species including P. falciparum. The results show PSNPs-MSP4/5 conjugates are highly immunogenic, inducing immune responses comprising both T helper 1 (Th1) and Th2 cellular immunity, and a spectrum of antibody subclasses including IgG1, IgG2a, and IgG2b. Benchmarked against Alum and Complete Freund's Adjuvant (CFA), the immune responses that were induced were of comparable or higher magnitude, for both T cell frequencies by ELISpot and antibody responses in terms of ELISA end titer. Importantly, immunization with PSNPs-MSP4/5 induced partial protection against malaria blood-stage infection (50-80%) shown to be mechanistically dependent on interferon gamma (IFN-γ) production. These results expand the scope of adjuvants considered for malaria blood-stage vaccine development to those that do not use conventional adjuvant pathways and emphasizes the critical role of cellular immunity and specifically IFN-γ producing cells in providing moderate protection against blood-stage malaria comparable to Freunds adjuvant.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Membrane Proteins/immunology , Nanoparticles/administration & dosage , Protozoan Proteins/immunology , Vaccines, Synthetic/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/immunology , Antibody Formation/immunology , Immunization/methods , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Th1 Cells/immunology , Th2 Cells/immunology
8.
Nanomaterials (Basel) ; 7(2)2017 Feb 10.
Article in English | MEDLINE | ID: mdl-28336871

ABSTRACT

DNA vaccines offer cost, flexibility, and stability advantages, but administered alone have limited immunogenicity. Previously, we identified optimal configurations of magnetic vectors comprising superparamagnetic iron oxide nanoparticles (SPIONs), polyethylenimine (PEI), and hyaluronic acid (HA) to deliver malaria DNA encoding Plasmodium yoelii (Py) merozoite surface protein MSP119 (SPIONs/PEI/DNA + HA gene complex) to dendritic cells and transfect them with high efficiency in vitro. Herein, we evaluate their immunogenicity in vivo by administering these potential vaccine complexes into BALB/c mice. The complexes induced antibodies against PyMSP119, with higher responses induced intraperitoneally than intramuscularly, and antibody levels further enhanced by applying an external magnetic field. The predominant IgG subclasses induced were IgG2a followed by IgG1 and IgG2b. The complexes further elicited high levels of interferon gamma (IFN-γ), and moderate levels of interleukin (IL)-4 and IL-17 antigen-specific splenocytes, indicating induction of T helper 1 (Th1), Th2, and Th17 cell mediated immunity. The ability of such DNA/nanoparticle complexes to induce cytophilic antibodies together with broad spectrum cellular immunity may benefit malaria vaccines.

9.
Oncotarget ; 8(3): 4062-4078, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27845897

ABSTRACT

The Polycomb repressive complex 2 (PRC2), which contains three core proteins EZH2, EED and SUZ12, controls chromatin compaction and transcription repression through trimethylation of lysine 27 on histone 3. The (7;17)(p15;q21) chromosomal translocation present in most cases of endometrial stromal sarcomas (ESSs) results in the in-frame fusion of the JAZF1 and SUZ12 genes. We have investigated whether and how the fusion protein JAZF1-SUZ12 functionally alters PRC2. We found that the fusion protein exists at high levels in ESS containing the t(7;17). Co-transient transfection assay indicated JAZF1-SUZ12 destabilized PRC2 components EZH2 and EED, resulting in decreased histone methyl transferase (HMT) activity, which was confirmed by in vitro studies using reconstituted PRC2 and nucleosome array substrates. We also demonstrated the PRC2 containing the fusion protein decreased the binding affinity to target chromatin loci. In addition, we found that trimethylation of H3K27 was decreased in ESS samples with the t(7;17), but there was no detectable change in H3K9 in these tissues. Moreover, re-expression of SUZ12 in Suz12 (-/-) ES cells rescued the neuronal differentiation while the fusion protein failed to restore this function and enhanced cell proliferation. In summary, our studies reveal that JAZF1-SUZ12 fusion protein disrupts the PRC2 complex, abolishes HMT activity and subsequently activates chromatin/genes normally repressed by PRC2. Such dyesfunction of PRC2 inhibits normal neural differentiation of ES cell and increases cell proliferation. Related changes induced by the JAZF-SUZ12 protein in endometrial stromal cells may explain the oncogenic effect of the t(7;17) in ESS.


Subject(s)
Chromatin/genetics , Endometrial Neoplasms/genetics , Neoplasm Proteins/genetics , Polycomb Repressive Complex 2/metabolism , Recombinant Fusion Proteins/genetics , Animals , Cell Differentiation , Cell Line , Chromatin Assembly and Disassembly , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 7/genetics , Co-Repressor Proteins , DNA-Binding Proteins , Epigenetic Repression , Female , HEK293 Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , Methylation , Mice , Polycomb Repressive Complex 2/genetics , Transcription Factors , Translocation, Genetic
10.
Mod Pathol ; 29(6): 570-81, 2016 06.
Article in English | MEDLINE | ID: mdl-26965583

ABSTRACT

A predominantly diffuse growth pattern and CD23 co-expression are uncommon findings in nodal follicular lymphoma and can create diagnostic challenges. A single case series in 2009 (Katzenberger et al) proposed a unique morphologic variant of nodal follicular lymphoma, characterized by a predominantly diffuse architecture, lack of the t(14;18) IGH/BCL2 translocation, presence of 1p36 deletion, frequent inguinal lymph node involvement, CD23 co-expression, and low clinical stage. Other studies on CD23+ follicular lymphoma, while associating inguinal location, have not specifically described this architecture. In addition, no follow-up studies have correlated the histopathologic and cytogenetic/molecular features of these cases, and they remain a diagnostic problem. We identified 11 cases of diffuse, CD23+ follicular lymphoma with histopathologic features similar to those described by Katzenberger et al. Along with pertinent clinical information, we detail their histopathology, IGH/BCL2 translocation status, lymphoma-associated chromosomal gains/losses, and assessment of mutations in 220 lymphoma-associated genes by massively parallel sequencing. All cases showed a diffuse growth pattern around well- to ill-defined residual germinal centers, uniform CD23 expression, mixed centrocytic/centroblastic cytology, and expression of at least one germinal center marker. Ten of 11 involved inguinal lymph nodes, 5 solely. By fluorescence in situ hybridization analysis, the vast majority lacked IGH/BCL2 translocation (9/11). Deletion of 1p36 was observed in five cases and included TNFRSF14. Of the six cases lacking 1p36 deletion, TNFRSF14 mutations were identified in three, highlighting the strong association of 1p36/TNFRSF14 abnormalities with this follicular lymphoma variant. In addition, 9 of the 11 cases tested (82%) had STAT6 mutations and nuclear P-STAT6 expression was detectable in the mutated cases by immunohistochemistry. The proportion of STAT6 mutations is higher than recently reported in conventional follicular lymphoma (11%). These findings lend support for a clinicopathologic variant of t(14;18) negative nodal follicular lymphoma and suggests importance of the interleukin (IL)-4/JAK/STAT6 pathway in this variant.


Subject(s)
Biomarkers, Tumor/genetics , Chromosome Disorders/genetics , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 18 , Lymphoma, Follicular/genetics , Mutation , Receptors, IgE/analysis , Receptors, Tumor Necrosis Factor, Member 14/genetics , STAT6 Transcription Factor/genetics , Translocation, Genetic , Adult , Aged, 80 and over , Biomarkers, Tumor/analysis , Chromosome Deletion , Chromosome Disorders/immunology , Chromosome Disorders/pathology , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 1/immunology , DNA Mutational Analysis/methods , Female , Genes, Immunoglobulin Heavy Chain , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lymphoma, Follicular/chemistry , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Male , Middle Aged , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , STAT6 Transcription Factor/analysis
11.
Leuk Lymphoma ; 57(4): 899-908, 2016.
Article in English | MEDLINE | ID: mdl-26294112

ABSTRACT

Genomic copy number alterations (CNAs) in diffuse large B-cell lymphoma (DLBCL) have roles in disease pathogenesis, but overall clinical relevance remains unclear. Herein, an unbiased algorithm was uniformly applied across three genome profiling datasets comprising 392 newly-diagnosed DLBCL specimens that defined 32 overlapping CNAs, involving 36 minimal common regions (MCRs). Scoring criteria were established for 50 aberrations within the MCRs while considering peak gains/losses. Application of these criteria to independent datasets revealed novel candidate genes with coordinated expression, such as CNOT2, potentially with pathogenic roles. No one single aberration significantly associated with patient outcome across datasets, but genomic complexity, defined by imbalance in more than one MCR, significantly portended adverse outcome in two of three independent datasets. Thus, the standardized scoring of CNAs currently developed can be uniformly applied across platforms, affording robust validation of genomic imbalance and complexity in DLBCL and overall clinical utility as biomarkers of patient outcome.


Subject(s)
Genetic Variation , Genomics , Lymphoma, Large B-Cell, Diffuse/genetics , Quantitative Trait Loci , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chromosome Aberrations , Comparative Genomic Hybridization , Cyclophosphamide/therapeutic use , DNA Copy Number Variations , Doxorubicin/therapeutic use , Female , Gene Expression Regulation, Neoplastic , Genetic Testing , Genomics/methods , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/mortality , Male , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prednisone/therapeutic use , Prognosis , Rituximab , Signal Transduction , Transcriptome , Vincristine/therapeutic use
12.
BMC Genomics ; 16: 676, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26335021

ABSTRACT

BACKGROUND: The metastasis-associated lung adenocarcinoma transcription 1 (Malat1) is a highly conserved long non-coding RNA (lncRNA) gene. Previous studies showed that Malat1 is abundantly expressed in many tissues and involves in promoting tumor growth and metastasis by modulating gene expression and target protein activities. However, little is known about the biological function and regulation mechanism of Malat1 in normal cell proliferation. RESULTS: In this study we conformed that Malat1 is highly conserved across vast evolutionary distances amongst 20 species of mammals in terms of sequence, and found that mouse Malat1 expresses in tissues of liver, kidney, lung, heart, testis, spleen and brain, but not in skeletal muscle. After treating erythroid myeloid lymphoid (EML) cells with All-trans Retinoic Acid (ATRA), we investigated the expression and regulation of Malat1 during hematopoietic differentiation, the results showed that ATRA significantly down regulates Malat1 expression during the differentiation of EML cells. Mouse LRH (Lin-Rhodamine(low) Hoechst(low)) cells that represent the early-stage progenitor cells show a high level of Malat1 expression, while LRB (Lin - Hoechst(Low) Rhodamine(Bright)) cells that represent the late-stage progenitor cells had no detectable expression of Malat1. Knockdown experiment showed that depletion of Malat1 inhibits the EML cell proliferation. Along with the down regulation of Malat1, the tumor suppressor gene p53 was up regulated during the differentiation. Interestingly, we found two p53 binding motifs with help of bioinformatic tools, and the following chromatin immunoprecipitation (ChIP) test conformed that p53 acts as a transcription repressor that binds to Malat1's promoter. Furthermore, we testified that p53 over expression in EML cells causes down regulation of Malat1. CONCLUSIONS: In summary, this study indicates Malat1 plays a critical role in maintaining the proliferation potential of early-stage hematopoietic cells. In addition to its biological function, the study also uncovers the regulation pattern of Malat1 expression mediated by p53 in hematopoietic differentiation. Our research shed a light on exploring the Malat1 biological role including therapeutic significance to inhibit the proliferation potential of malignant cells.


Subject(s)
Cell Differentiation , Conserved Sequence/genetics , Evolution, Molecular , Hematopoiesis , RNA, Long Noncoding/genetics , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Differentiation/drug effects , Cell Proliferation , Down-Regulation/genetics , Hematopoiesis/drug effects , Humans , Liver/drug effects , Liver/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice, Inbred BALB C , Primates , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Long Noncoding/metabolism , Species Specificity , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
13.
J Urol ; 193(5): 1479-85, 2015 May.
Article in English | MEDLINE | ID: mdl-25498568

ABSTRACT

PURPOSE: Accurate discrimination of benign oncocytoma and malignant renal cell carcinoma is useful for planning appropriate treatment strategies for patients with renal masses. Classification of renal neoplasms solely based on histopathology can be challenging, especially the distinction between chromophobe renal cell carcinoma and oncocytoma. In this study we develop and validate an algorithm based on genomic alterations for the classification of common renal neoplasms. MATERIALS AND METHODS: Using TCGA renal cell carcinoma copy number profiles and the published literature, a classification algorithm was developed and scoring criteria were established for the presence of each genomic marker. As validation, 191 surgically resected formalin fixed paraffin embedded renal neoplasms were blindly submitted to targeted array comparative genomic hybridization and classified according to the algorithm. CCND1 rearrangement was assessed by fluorescence in situ hybridization. RESULTS: The optimal classification algorithm comprised 15 genomic markers, and involved loss of VHL, 3p21 and 8p, and chromosomes 1, 2, 6, 10 and 17, and gain of 5qter, 16p, 17q and 20q, and chromosomes 3, 7 and 12. On histological rereview (leading to the exclusion of 3 specimens) and using histology as the gold standard, 58 of 62 (93%) clear cell, 51 of 56 (91%) papillary and 33 of 34 (97%) chromophobe renal cell carcinomas were classified correctly. Of the 36 oncocytoma specimens 33 were classified as oncocytoma (17 by array comparative genomic hybridization and 10 by array comparative genomic hybridization plus fluorescence in situ hybridization) or benign (6). Overall 93% diagnostic sensitivity and 97% specificity were achieved. CONCLUSIONS: In a clinical diagnostic setting the implementation of genome based molecular classification could serve as an ancillary assay to assist in the histological classification of common renal neoplasms.


Subject(s)
Adenoma, Oxyphilic/classification , Adenoma, Oxyphilic/genetics , Algorithms , Carcinoma, Renal Cell/classification , Carcinoma, Renal Cell/genetics , Genomics , Kidney Cortex , Kidney Neoplasms/classification , Kidney Neoplasms/genetics , Comparative Genomic Hybridization , Humans , In Situ Hybridization, Fluorescence
14.
Methods Mol Biol ; 1143: 181-94, 2014.
Article in English | MEDLINE | ID: mdl-24715289

ABSTRACT

The efficiency of delivery of DNA vaccines is often relatively low compared to protein vaccines. The use of superparamagnetic iron oxide nanoparticles (SPIONs) to deliver genes via magnetofection shows promise in improving the efficiency of gene delivery both in vitro and in vivo. In particular, the duration for gene transfection especially for in vitro application can be significantly reduced by magnetofection compared to the time required to achieve high gene transfection with standard protocols. SPIONs that have been rendered stable in physiological conditions can be used as both therapeutic and diagnostic agents due to their unique magnetic characteristics. Valuable features of iron oxide nanoparticles in bioapplications include a tight control over their size distribution, magnetic properties of these particles, and the ability to carry particular biomolecules to specific targets. The internalization and half-life of the particles within the body depend upon the method of synthesis. Numerous synthesis methods have been used to produce magnetic nanoparticles for bioapplications with different sizes and surface charges. The most common method for synthesizing nanometer-sized magnetite Fe3O4 particles in solution is by chemical coprecipitation of iron salts. The coprecipitation method is an effective technique for preparing a stable aqueous dispersions of iron oxide nanoparticles. We describe the production of Fe3O4-based SPIONs with high magnetization values (70 emu/g) under 15 kOe of the applied magnetic field at room temperature, with 0.01 emu/g remanence via a coprecipitation method in the presence of trisodium citrate as a stabilizer. Naked SPIONs often lack sufficient stability, hydrophilicity, and the capacity to be functionalized. In order to overcome these limitations, polycationic polymer was anchored on the surface of freshly prepared SPIONs by a direct electrostatic attraction between the negatively charged SPIONs (due to the presence of carboxylic groups) and the positively charged polymer. Polyethylenimine was chosen to modify the surface of SPIONs to assist the delivery of plasmid DNA into mammalian cells due to the polymer's extensive buffering capacity through the "proton sponge" effect.


Subject(s)
Magnetite Nanoparticles , Vaccination/methods , Vaccines, DNA/administration & dosage , Animals , Cell Line , Ferric Compounds/chemistry , Gene Transfer Techniques , Humans , Magnetite Nanoparticles/chemistry , Polyethyleneimine/chemistry , Vaccines, DNA/genetics , Vaccines, DNA/immunology
15.
Appl Microbiol Biotechnol ; 98(8): 3669-78, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24146077

ABSTRACT

Plasmodium falciparum is the causative agent of the most serious form of malaria. Although a combination of control measures has significantly limited malaria morbidity and mortality in the last few years, it is generally agreed that sustained control or even eradication will require additional tools including an effective malaria vaccine. Merozoite surface protein 4, MSP4, which is present during the asexual stage of P. falciparum, is a recognized target that would be useful in a subunit vaccine against blood stages of malaria. Falciparum malaria is most prevalent in developing countries, and this in turn leads to a requirement for safe, low-cost vaccines. We have attempted to utilize the nonpathogenic, gram-positive organism Bacillus subtilis to produce PfMSP4. PfMSP4 was secreted into the culture medium at a yield of 4.5 mg/L. Characterization studies including SDS-PAGE, mass spectrometry, and N-terminal sequencing indicated that the B. subtilis expression system secreted a full length PfMSP4 protein compared to a truncated version in Escherichia coli. Equivalent amounts of purified B. subtilis and E. coli-derived PfMSP4 were used for immunization studies, resulting in statistically significant higher mean titer values for the B. subtilis-derived immunogen. The mouse antibodies raised against B. subtilis produced PfMSP4 that were reactive to parasite proteins as evidenced by immunoblotting on parasite lysate and indirect immunofluorescence assays of fixed parasites. The B. subtilis expression system, in contrast to E. coli, expresses higher amounts of full length PfMSP4 products, decreased levels of aggregates, and allows the development of simplified downstream processing procedures.


Subject(s)
Antigens, Protozoan/immunology , Antigens, Protozoan/isolation & purification , Bacillus subtilis/genetics , Malaria Vaccines/immunology , Malaria Vaccines/isolation & purification , Protozoan Proteins/immunology , Protozoan Proteins/isolation & purification , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Immunoblotting , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mass Spectrometry , Mice , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Sequence Analysis, Protein , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification
16.
J Control Release ; 168(1): 10-7, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23500060

ABSTRACT

We investigated the efficacy and types of immune responses from plasmid malaria DNA vaccine encoding VR1020-PyMSP119 condensed on the surface of polyethyleneimine (PEI)-coated SPIONs. In vivo mouse studies were done firstly to determine the optimum magnetic vector composition, and then to observe immune responses elicited when magnetic vectors were introduced via different administration routes. Higher serum antibody titers against PyMSP119 were observed with intraperitoneal and intramuscular injections than subcutaneous and intradermal injections. Robust IgG2a and IgG1 responses were observed for intraperitoneal administration, which could be due to the physiology of peritoneum as a major reservoir of macrophages and dendritic cells. Heterologous DNA prime followed by single protein boost vaccination regime also enhanced IgG2a, IgG1, and IgG2b responses, indicating the induction of appropriate memory immunity that can be elicited by protein on recall. These outcomes support the possibility to design superparamagnetic nanoparticle-based DNA vaccines to optimally evoke desired antibody responses, useful for a variety of diseases including malaria.


Subject(s)
Ferric Compounds/chemistry , Malaria Vaccines/administration & dosage , Nanoparticles/administration & dosage , Polyethyleneimine/chemistry , Vaccines, DNA/administration & dosage , Animals , Antibodies, Protozoan/blood , DNA/administration & dosage , DNA/chemistry , Female , Genetic Vectors , Immunoglobulin G/blood , Magnetic Phenomena , Malaria Vaccines/chemistry , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Vaccines, DNA/chemistry
17.
Parasitol Int ; 62(2): 134-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23183102

ABSTRACT

The bioinformatics software, Geneious, provides a useful platform for researchers to retrieve and analyse genomic and functional genomics information. However, the main databases that the software is able to access are hosted by NCBI (National Center for Biotechnology Information). The databases of EuPathDB (Eukaryotic Pathogen Database Resources), such as PlasmoDB and PiroplasmaDB, collect more specific and detailed information about eukaryotic pathogens than those kept in NCBI databases. Two plugins for Geneious, one for PlasmaDB and one for PiroplasmaDB were developed. When installed, users can use search facilities to find and import gene and protein sequences from the EuPathDB databases. Users can then use the functions of Geneious to process the sequence information. When information unique to PlasmoDB and PiroplasmaDB is required, the user can access results linked with the gene/protein sequence via the default web browser. The plugins are freely available from the Victorian Bioinformatics Consortium website. The plugins can be modified to access any of the databases of EuPathDB.


Subject(s)
Computational Biology/methods , Databases, Genetic , Genome, Protozoan/genetics , Piroplasmida/genetics , Plasmodium/genetics , Software , Animals , Internet , Systems Integration , User-Computer Interface
18.
Appl Microbiol Biotechnol ; 94(1): 151-61, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22170105

ABSTRACT

Development of a safe, effective and affordable malaria vaccine is central to global disease control efforts. One of the most highly regarded proteins for inclusion in an asexual blood stage subunit vaccine is the 19-kDa C-terminal fragment of merozoite surface protein 1 (MSP1(19)). As production of vaccine antigens in plants can potentially overcome cost and delivery hurdles, we set out to produce MSP1(19) in plants, characterise the protein and test its immunogenicity using a mouse model. Plasmodium yoelii MSP1(19) (PyMSP1(19)) was produced in Nicotiana benthamiana using the MagnICON® deconstructed TMV-based viral vector. PyMSP1(19) yield of at least 23% total soluble protein (TSP;3-4 mg/g Fwt) were achieved using a codon-optimised construct that was targeted to the apoplast. Freeze-dried leaf powder contained at least 20 mg PyMSP1(19) per gram dry weight and the protein retained immunogenicity in this form for more than 2 years. Characterisation studies, including SDS-PAGE, mass spectrometry and circular dichroism, indicated that the plant-expressed PyMSP1(19) was similar to its Escherichia coli- and Saccharomyces cerevisiae-expressed counterparts. Purified plant-made PyMSP1(19) induced strong immune responses following intraperitoneal immunisation, although titres were lower than those induced by an equivalent dose of purified E. coli-expressed PyMSP1(19). The reason for this is uncertain but may be due to differences in the oligomerisation profile of the vaccines. The plant-made PyMSP1(19) vaccine was also found to be orally immunogenic when delivered alone or following immunisation with a PyMSP1(19) DNA vaccine. This study adds to an increasing body of research supporting the feasibility of plants as both a factory for the production of malaria antigens, and as a safe and affordable platform for oral delivery of a temperature-stable malaria vaccine.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Malaria/immunology , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/immunology , Nicotiana/genetics , Plasmodium yoelii/immunology , Amino Acid Motifs , Animals , Antigens, Protozoan/chemistry , Female , Gene Expression , Humans , Immunization , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Merozoite Surface Protein 1/chemistry , Mice , Mice, Inbred BALB C , Plasmodium yoelii/chemistry , Plasmodium yoelii/genetics , Plasmodium yoelii/growth & development , Nicotiana/metabolism
19.
Proc Natl Acad Sci U S A ; 108(10): 3982-7, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21368137

ABSTRACT

Microtubules are integral to neuronal development and function. They endow cells with polarity, shape, and structure, and their extensive surface area provides substrates for intracellular trafficking and scaffolds for signaling molecules. Consequently, microtubule polymerization dynamics affect not only structural features of the cell but also the subcellular localization of proteins that can trigger intracellular signaling events. In the nematode Caenorhabditis elegans, the processes of touch receptor neurons are filled with a bundle of specialized large-diameter microtubules. We find that conditions that disrupt these microtubules (loss of either the MEC-7 ß-tubulin or MEC-12 α-tubulin or growth in 1 mM colchicine) cause a general reduction in touch receptor neuron (TRN) protein levels. This reduction requires a p38 MAPK pathway (DLK-1, MKK-4, and PMK-3) and the transcription factor CEBP-1. Cells may use this feedback pathway that couples microtubule state and MAPK activation to regulate cellular functions.


Subject(s)
Caenorhabditis elegans/metabolism , Gene Expression , Microtubules/metabolism , Neurons/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Colchicine/pharmacology , Mutation , Neurons/drug effects , Neurons/enzymology
20.
Langmuir ; 27(7): 3703-12, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21361304

ABSTRACT

Low efficiency is often observed in the delivery of DNA vaccines. The use of superparamagnetic nanoparticles (SPIONs) to deliver genes via magnetofection could improve transfection efficiency and target the vector to its desired locality. Here, magnetofection was used to enhance the delivery of a malaria DNA vaccine encoding Plasmodium yoelii merozoite surface protein MSP1(19) (VR1020-PyMSP1(19)) that plays a critical role in Plasmodium immunity. The plasmid DNA (pDNA) containing membrane associated 19-kDa carboxyl-terminal fragment of merozoite surface protein 1 (PyMSP1(19)) was conjugated with superparamagnetic nanoparticles coated with polyethyleneimine (PEI) polymer, with different molar ratio of PEI nitrogen to DNA phosphate. We reported the effects of SPIONs-PEI complexation pH values on the properties of the resulting particles, including their ability to condense DNA and the gene expression in vitro. By initially lowering the pH value of SPIONs-PEI complexes to 2.0, the size of the complexes decreased since PEI contained a large number of amino groups that became increasingly protonated under acidic condition, with the electrostatic repulsion inducing less aggregation. Further reaggregation was prevented when the pHs of the complexes were increased to 4.0 and 7.0, respectively, before DNA addition. SPIONs/PEI complexes at pH 4.0 showed better binding capability with PyMSP1(19) gene-containing pDNA than those at neutral pH, despite the negligible differences in the size and surface charge of the complexes. This study indicated that the ability to protect DNA molecules due to the structure of the polymer at acidic pH could help improve the transfection efficiency. The transfection efficiency of magnetic nanoparticle as carrier for malaria DNA vaccine in vitro into eukaryotic cells, as indicated via PyMSP1(19) expression, was significantly enhanced under the application of external magnetic field, while the cytotoxicity was comparable to the benchmark nonviral reagent (Lipofectamine 2000).


Subject(s)
Malaria Vaccines/chemistry , Nanoparticles/chemistry , Polyethyleneimine/chemistry , Vaccines, DNA/chemistry , Animals , COS Cells , Cell Survival/drug effects , Chlorocebus aethiops , Drug Carriers/administration & dosage , Drug Carriers/adverse effects , Drug Carriers/chemistry , Malaria Vaccines/administration & dosage , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nanoparticles/administration & dosage , Nanoparticles/adverse effects , Nanoparticles/ultrastructure , Nanotechnology , Vaccines, DNA/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...