Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Immunology ; 171(2): 235-249, 2024 Feb.
Article En | MEDLINE | ID: mdl-37947218

The incidence of systemic lupus erythematosus (SLE) is about nine times higher in women than in men, and the underlying mechanisms that contribute to this gender bias are not fully understood. Previously, using lupus-prone (SWR × NZB)F1 (SNF1) mice, we have shown that the intestinal immune system could play a role in the initiation and progression of disease in SLE, and depletion of gut microbiota produces more pronounced disease protection in females than in males. Here, we show that the gut permeability features of lupus-prone female SNF1 mice at juvenile ages directly correlate with the expression levels of pro-inflammatory factors, faecal IgA abundance and nAg reactivity and the eventual systemic autoantibody levels and proteinuria onset. Furthermore, we observed that the disease protection achieved in female SNF1 mice upon depletion of gut microbiota correlates with the diminished gut inflammatory protein levels, intestinal permeability and circulating microbial DNA levels. However, faecal microbiota transplant from juvenile male and females did not result in modulation of gut inflammatory features or permeability. Overall, these observations suggest that the early onset of intestinal inflammation, systemic autoantibody production and clinical stage disease in lupus-prone females is linked to higher gut permeability in them starting at as early as juvenile age. While the higher gut permeability in juvenile lupus-prone females is dependent on the presence of gut microbes, it appears to be independent of the composition of gut microbiota.


Autoimmunity , Lupus Erythematosus, Systemic , Female , Humans , Male , Mice , Animals , Intestinal Barrier Function , Sexism , Mice, Inbred NZB , Autoantibodies , Disease Models, Animal
2.
Autoimmunity ; 55(2): 95-108, 2022 03.
Article En | MEDLINE | ID: mdl-34882054

Progressive destruction of pancreatic islet ß-cells by immune cells is a primary feature of type 1 diabetes (T1D) and therapies that can restore the functional ß-cell mass are needed to alleviate disease progression. Here, we report the use of mesenchymal stromal/stem cells (MSCs) for the production and delivery of Gastrin, a peptide hormone that is produced by intestinal cells and foetal islets and can increase ß-Cell mass, to promote protection from T1D. A single injection of syngeneic MSCs that were engineered to express Gastrin (Gastrin-MSCs) caused a significant delay in hyperglycaemia in non-obese diabetic (NOD) mice compared to engineered control-MSCs. Similar treatment of early-hyperglycaemic mice caused the restoration of euglycemia for a considerable duration, and these therapeutic effects were associated with the protection of, and/or higher frequencies of, insulin-producing islets and less severe insulitis. While the overall immune cell phenotype was not affected profoundly upon treatment using Gastrin-MSCs or upon in vitro culture, pancreatic lymph node cells from Gastrin-MSC treated mice, upon ex vivo challenge with self-antigen, showed a Th2 and Th17 bias, and diminished the diabetogenic property in NOD-Rag1 deficient mice suggesting a disease protective immune modulation under Gastrin-MSC treatment associated protection from hyperglycaemia. Overall, this study shows the potential of production and delivery of Gastrin in vivo, by MSCs, in protecting insulin-producing ß-cells and ameliorating the disease progression in T1D.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Gastrins , Islets of Langerhans , Mesenchymal Stem Cell Transplantation , Animals , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Gastrins/genetics , Mesenchymal Stem Cells , Mice , Mice, Inbred NOD
3.
J Autoimmun ; 108: 102420, 2020 03.
Article En | MEDLINE | ID: mdl-32019684

The risk of developing systemic lupus erythematosus (SLE) is about 9 times higher in women as compared to men. Our recent report, which used (SWRxNZB) F1 (SNF1) mouse model of spontaneous lupus, showed a potential link between immune response initiated in the gut mucosa at juvenile age (sex hormone independent) and SLE susceptibility. Here, using this mouse model, we show that gut microbiota contributes differently to pro-inflammatory immune response in the intestine and autoimmune progression in lupus-prone males and females. We found that gut microbiota composition in male and female littermates are significantly different only at adult ages. However, depletion of gut microbes causes suppression of autoimmune progression only in females. In agreement, microbiota depletion suppressed the pro-inflammatory cytokine response of gut mucosa in juvenile and adult females. Nevertheless, microbiota from females and males showed, upon cross-transfer, contrasting abilities to modulate disease progression. Furthermore, orchidectomy (castration) not only caused changes in the composition of gut microbiota, but also a modest acceleration of autoimmune progression. Overall, our work shows that microbiota-dependent pro-inflammatory immune response in the gut mucosa of females initiated at juvenile ages and androgen-dependent protection of males contribute to gender differences in the intestinal immune phenotype and systemic autoimmune progression.


Autoimmunity , Gastrointestinal Microbiome/immunology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/pathology , Phenotype , Age Factors , Animals , Biomarkers , Disease Models, Animal , Disease Progression , Female , Inflammation Mediators/metabolism , Male , Metagenome , Metagenomics/methods , Mice , Sex Factors , Transcriptome
4.
Diabetes ; 68(10): 1975-1989, 2019 10.
Article En | MEDLINE | ID: mdl-31311801

Bacteroides fragilis (BF) is an integral component of the human colonic commensal microbiota. BF is also the most commonly isolated organism from clinical cases of intra-abdominal abscesses, suggesting its potential to induce proinflammatory responses upon accessing the systemic compartment. Hence, we examined the impact of mucosal and systemic exposures to BF on type 1 diabetes (T1D) incidence in NOD mice. The impact of intestinal exposure to BF under a chemically induced enhanced gut permeability condition, which permits microbial translocation, in T1D was also examined. While oral administration of heat-killed (HK) BF to prediabetic mice caused enhanced immune regulation and suppression of autoimmunity, resulting in delayed hyperglycemia, mice that received HK BF by intravenous injection showed rapid disease progression. Importantly, polysaccharide A-deficient BF failed to produce these opposing effects upon oral and systemic deliveries. Furthermore, BF-induced modulation of disease progression was observed in wild-type, but not TLR2-deficient, NOD mice. Interestingly, oral administration of BF under enhanced gut permeability conditions resulted in accelerated disease progression and rapid onset of hyperglycemia in NOD mice. Overall, these observations suggest that BF-like gut commensals can cause proinflammatory responses upon gaining access to the systemic compartment and contribute to T1D in at-risk subjects.


Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Animals , Autoimmunity/immunology , Bacteroides fragilis , Disease Progression , Humans , Mice , Polysaccharides
5.
Sci Transl Med ; 10(431)2018 03 07.
Article En | MEDLINE | ID: mdl-29514998

Systemic sclerosis (SSc) is a debilitating inflammatory and fibrotic disease that affects the skin and internal organs. Although the pathophysiology of SSc remains poorly characterized, mononuclear cells, mainly macrophages and T cells, have been implicated in inflammation and fibrosis. Inducible costimulator (ICOS), which is expressed on a subset of memory T helper (TH) and T follicular helper (TFH) cells, has been shown to be increased in SSc and associated with disease pathology. However, the identity of the relevant ICOS+ T cells and their contribution to inflammation and fibrosis in SSc are still unknown. We show that CD4+ ICOS-expressing T cells with a TFH-like phenotype infiltrate the skin of patients with SSc and are correlated with dermal fibrosis and clinical disease status. ICOS+ TFH-like cells were found to be increased in the skin of graft-versus-host disease (GVHD)-SSc mice and contributed to dermal fibrosis via an interleukin-21- and matrix metalloproteinase 12-dependent mechanism. Administration of an anti-ICOS antibody to GVHD-SSc mice prevented the expansion of ICOS+ TFH-like cells and inhibited inflammation and dermal fibrosis. Interleukin-21 neutralization in GVHD-SSc mice blocked disease pathogenesis by reducing skin fibrosis. These results identify ICOS+ TFH-like profibrotic cells as key drivers of fibrosis in a GVHD-SSc model and suggest that inhibition of these cells could offer therapeutic benefit for SSc.


Fibrosis/immunology , Fibrosis/metabolism , Scleroderma, Systemic/immunology , Scleroderma, Systemic/metabolism , T-Lymphocytes/metabolism , Animals , Female , Fibrosis/therapy , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Graft vs Host Disease/therapy , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukins/antagonists & inhibitors , Interleukins/metabolism , Mice , Mice, Inbred BALB C , Receptors, Interleukin-21/metabolism , Scleroderma, Systemic/therapy , Skin/immunology , Skin/metabolism , Skin Diseases/immunology , Skin Diseases/metabolism , Skin Diseases/therapy
6.
Sci Rep ; 6: 27379, 2016 06 07.
Article En | MEDLINE | ID: mdl-27271479

The proliferation and survival of hematopoietic stem cells (HSCs) has to be strictly coordinated to ensure the timely production of all blood cells. Here we report that the splice factor and RNA binding protein hnRNP L (heterogeneous nuclear ribonucleoprotein L) is required for hematopoiesis, since its genetic ablation in mice reduces almost all blood cell lineages and causes premature death of the animals. In agreement with this, we observed that hnRNP L deficient HSCs lack both the ability to self-renew and foster hematopoietic differentiation in transplanted hosts. They also display mitochondrial dysfunction, elevated levels of γH2AX, are Annexin V positive and incorporate propidium iodide indicating that they undergo cell death. Lin(-)c-Kit(+) fetal liver cells from hnRNP L deficient mice show high p53 protein levels and up-regulation of p53 target genes. In addition, cells lacking hnRNP L up-regulated the expression of the death receptors TrailR2 and CD95/Fas and show Caspase-3, Caspase-8 and Parp cleavage. Treatment with the pan-caspase inhibitor Z-VAD-fmk, but not the deletion of p53, restored cell survival in hnRNP L deficient cells. Our data suggest that hnRNP L is critical for the survival and functional integrity of HSCs by restricting the activation of caspase-dependent death receptor pathways.


Cell Survival/physiology , Hematopoietic Stem Cells/cytology , Heterogeneous-Nuclear Ribonucleoprotein L/physiology , Animals , Apoptosis/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Stress, Physiological , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
PLoS One ; 10(5): e0125998, 2015.
Article En | MEDLINE | ID: mdl-25993117

Alternative splicing patterns are known to vary between tissues but these patterns have been found to be predominantly peculiar to one species or another, implying only a limited function in fundamental neural biology. Here we used high-throughput RT-PCR to monitor the expression pattern of all the annotated simple alternative splicing events (ASEs) in the Reference Sequence Database, in different mouse tissues and identified 93 brain-specific events that shift from one isoform to another (switch-like) between brain and other tissues. Consistent with an important function, regulation of a core set of 9 conserved switch-like ASEs is highly conserved, as they have the same pattern of tissue-specific splicing in all vertebrates tested: human, mouse and zebrafish. Several of these ASEs are embedded within genes that encode proteins associated with the neuronal microtubule network, and show a dramatic and concerted shift within a short time window of human neural stem cell differentiation. Similarly these exons are dynamically regulated in zebrafish development. These data demonstrate that although alternative splicing patterns often vary between species, there is nonetheless a core set of vertebrate brain-specific ASEs that are conserved between species and associated with neural differentiation.


Alternative Splicing , Cell Differentiation , Neurons/cytology , Vertebrates/metabolism , Animals , Humans , Mice , Real-Time Polymerase Chain Reaction , Zebrafish
8.
Cancer Cell ; 23(2): 200-14, 2013 Feb 11.
Article En | MEDLINE | ID: mdl-23410974

Most patients with acute lymphoblastic leukemia (ALL) fail current treatments highlighting the need for better therapies. Because oncogenic signaling activates a p53-dependent DNA damage response and apoptosis, leukemic cells must devise appropriate countermeasures. We show here that growth factor independence 1 (Gfi1) can serve such a function because Gfi1 ablation exacerbates p53 responses and lowers the threshold for p53-induced cell death. Specifically, Gfi1 restricts p53 activity and expression of proapoptotic p53 targets such as Bax, Noxa (Pmaip1), and Puma (Bbc3). Subsequently, Gfi1 ablation cures mice from leukemia and limits the expansion of primary human T-ALL xenografts in mice. This suggests that targeting Gfi1 could improve the prognosis of patients with T-ALL or other lymphoid leukemias.


Apoptosis , DNA Damage/genetics , DNA-Binding Proteins/physiology , Lymphoma, B-Cell/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , Animals , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, Notch1/genetics , Xenograft Model Antitumor Assays
9.
Blood ; 120(19): 4006-17, 2012 Nov 08.
Article En | MEDLINE | ID: mdl-22932805

The coding single nucleotide polymorphism GFI136N in the human gene growth factor independence 1 (GFI1) is present in 3%-7% of whites and increases the risk for acute myeloid leukemia (AML) by 60%. We show here that GFI136N, in contrast to GFI136S, lacks the ability to bind to the Gfi1 target gene that encodes the leukemia-associated transcription factor Hoxa9 and fails to initiate histone modifications that regulate HoxA9 expression. Consistent with this, AML patients heterozygous for the GFI136N variant show increased HOXA9 expression compared with normal controls. Using ChipSeq, we demonstrate that GFI136N specific epigenetic changes are also present in other genes involved in the development of AML. Moreover, granulomonocytic progenitors, a bone marrow subset from which AML can arise in humans and mice, show a proliferative expansion in the presence of the GFI136N variant. In addition, granulomonocytic progenitors carrying the GFI136N variant allele have altered gene expression patterns and differ in their ability to grow after transplantation. Finally, GFI136N can accelerate a K-RAS driven fatal myeloproliferative disease in mice. Our data suggest that the presence of a GFI136N variant allele induces a preleukemic state in myeloid precursors by deregulating the expression of Hoxa9 and other AML-related genes.


DNA-Binding Proteins/genetics , Epigenesis, Genetic , Homeodomain Proteins/genetics , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics , Animals , Cluster Analysis , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Hematopoiesis/genetics , Histones/metabolism , Humans , Mice , Mice, Transgenic , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/mortality , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/metabolism
10.
J Immunol ; 188(11): 5377-88, 2012 Jun 01.
Article En | MEDLINE | ID: mdl-22523384

The regulation of posttranscriptional modifications of pre-mRNA by alternative splicing is important for cellular function, development, and immunity. The receptor tyrosine phosphatase CD45, which is expressed on all hematopoietic cells, is known for its role in the development and activation of T cells. CD45 is known to be alternatively spliced, a process that is partially regulated by heterogeneous nuclear ribonucleoprotein (hnRNP) L. To investigate the role of hnRNP L further, we have generated conditional hnRNP L knockout mice and found that LckCre-mediated deletion of hnRNP L results in a decreased thymic cellularity caused by a partial block at the transition stage between double-negative 4 and double-positive cells. In addition, hnRNP L(-/-) thymocytes express aberrant levels of the CD45RA splice isoforms and show high levels of phosphorylated Lck at the activator tyrosine Y394, but lack phosphorylation of the inhibitory tyrosine Y505. This indicated an increased basal Lck activity and correlated with higher proliferation rates of double-negative 4 cells in hnRNP L(-/-) mice. Deletion of hnRNP L also blocked the migration and egress of single-positive thymocytes to peripheral lymphoid organs in response to sphingosine-1-phosphate and the chemokines CCL21 and CXCL12 very likely as a result of aberrant splicing of genes encoding GTPase regulators and proteins affecting cytoskeletal organization. Our results indicate that hnRNP L regulates T cell differentiation and migration by regulating pre-TCR and chemokine receptor signaling.


Alternative Splicing/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Cell Proliferation , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Stem Cells/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Alternative Splicing/genetics , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
11.
Stem Cells ; 29(2): 376-85, 2011 Feb.
Article En | MEDLINE | ID: mdl-21732494

The regulation of gene transcription is elementary for the function of hematopoietic stem cells (HSCs). The transcriptional repressor growth factor independence 1 (Gfi1) restricts HSC proliferation and is essential to maintain their self-renewal capacity and multipotency after transplantation. In addition, Gfi1(-/-) HSCs are severely compromised in their ability to compete with wild-type (wt) HSCs after transplantation. We now report that Gfi1 protects HSCs against stress-induced apoptosis, probably, by repressing the proapoptotic target gene Bax, since irradiated Gfi1(-/-) HSCs display higher expression of Bax and show a higher rate of apoptosis than wt HSCs. This protective function of Gfi1 appears to be functionally relevant since Gfi1(-/-) HSCs that express Bcl-2, which antagonizes the effects of Bax, regain their ability to self renew and to initiate multilineage differentiation after transplantation. Surprisingly, Gfi1(-/-) xBcl-2 transgenic mice also show a strong, systemic expansion of Mac-1(+) Gr-1(-) myeloid cells in bone marrow and peripheral lymphoid organs. These cells express high levels of the proleukemogenic transcription factor Hoxa9 and, in older mice, appear as atypical monocytoid-blastoid cells in the peripheral blood. As a result of this massive expansion of myeloid cells, all Gfi1(-/-) xBcl-2 mice eventually succumb to a myeloproliferative-like disease resembling a preleukemic state. In summary, our data demonstrate that Gfi1's ability to protect against apoptosis is essential for HSC function. In addition, our finding show that Gfi1 prevents the development of myeloproliferative diseases and provides evidence how Gfi1 deficiency could be linked to myeloid leukemia.


Apoptosis/genetics , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Myeloproliferative Disorders/prevention & control , Transcription Factors/metabolism , Animals , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/deficiency , Gene Knockout Techniques , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Myeloid Cells , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transcription Factors/deficiency , bcl-2-Associated X Protein/antagonists & inhibitors
12.
Blood ; 116(24): 5149-61, 2010 Dec 09.
Article En | MEDLINE | ID: mdl-20826720

Donor-matched transplantation of hematopoietic stem cells (HSCs) is widely used to treat hematologic malignancies but is associated with high mortality. The expansion of HSC numbers and their mobilization into the bloodstream could significantly improve therapy. We report here that adult mice conditionally deficient for the transcription Growth factor independence 1b (Gfi1b) show a significant expansion of functional HSCs in the bone marrow and blood. Despite this expansion, Gfi1b(ko/ko) HSCs retain their ability to self-renew and to initiate multilineage differentiation but are no longer quiescent and contain elevated levels of reactive oxygen species. Treatment of Gfi1b(ko/ko) mice with N-acetyl-cystein significantly reduced HSC numbers indicating that increased reactive oxygen species levels are at least partially responsible for the expansion of Gfi1b-deficient HSCs. Moreover, Gfi1b(-/-) HSCs show decreased expression of CXCR4 and Vascular cell adhesion protein-1, which are required to retain dormant HSCs in the endosteal niche, suggesting that Gfi1b regulates HSC dormancy and pool size without affecting their function. Finally, the additional deletion of the related Gfi1 gene in Gfi1b(ko/ko) HSCs is incompatible with the maintenance of HSCs, suggesting that Gfi1b and Gfi1 have partially overlapping functions but that at least one Gfi gene is essential for the generation of HSCs.


Cell Movement , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Acetylcysteine/pharmacology , Amine Oxidase (Copper-Containing)/biosynthesis , Animals , Cell Adhesion Molecules/biosynthesis , DNA-Binding Proteins/physiology , Homeostasis , Mice , Mice, Knockout , Proto-Oncogene Proteins/deficiency , Reactive Oxygen Species , Receptors, CXCR4/biosynthesis , Repressor Proteins/deficiency , Transcription Factors/physiology
13.
Mol Cell Biol ; 30(16): 3929-42, 2010 Aug.
Article En | MEDLINE | ID: mdl-20547752

Endotoxin (bacterial lipopolysaccharide [LPS]) causes fatal septic shock via the Toll-like receptor 4 (TLR-4) protein present on innate immunity effector cells, which activates nuclear factor kappa B (NF-kappaB), inducing proinflammatory cytokines, including tumor necrosis factor alpha (TNF-alpha). An early step in this process involves nuclear sequestration of the p65-RelA NF-kappaB subunit, enabling transcriptional activation of target inflammatory cytokine genes. Here, we analyzed the role of the nuclear zinc finger protein Gfi1 in the TLR response using primary bone marrow-derived macrophages. We show that upon LPS stimulation, expression of Gfi1 is induced with kinetics similar to those of nuclear translocation of p65 and that Gfi1 interacts with p65 and inhibits p65-mediated transcriptional transactivation by interfering with p65 binding to target gene promoter DNA. Gfi1-deficient macrophages show abnormally high mRNA levels of the TNF-alpha gene and many other p65 target genes and a higher rate of TNF promoter occupancy by p65 than wild-type cells after LPS stimulation, suggesting that Gfi1 functions as an antagonist of NF-kappaB activity at the level of promoter binding. Our findings identify a new function of Gfi1 as a general negative regulator of the endotoxin-initiated innate immune responses, including septic shock and possibly other severe inflammatory diseases.


DNA-Binding Proteins/metabolism , Inflammation/immunology , Inflammation/metabolism , Toll-Like Receptors/metabolism , Transcription Factor RelA/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , DNA/genetics , DNA/metabolism , DNA Primers/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , Immunity, Innate/drug effects , Inflammation/etiology , Lipopolysaccharides/toxicity , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Shock, Septic/etiology , Shock, Septic/immunology , Shock, Septic/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency , Transcription Factors/genetics , Transcriptional Activation , Tumor Necrosis Factor-alpha/genetics , Zinc Fingers
14.
Vaccine ; 25(5): 814-24, 2007 Jan 15.
Article En | MEDLINE | ID: mdl-17027124

To investigate the strategy of using a multivalent polyprotein DNA vaccine against Staphylococcus aureus, a series of plasmids was used to immunize mice followed by infectious challenge. The plasmid vaccines expressed Clumping factor A (Clfa), fibronectin binding protein A (FnBPA) and the enzyme Sortase (Srt) as single proteins or combined as a polyprotein. All animals produced a mixed Th1 and Th2 response including functional antigen-specific, mostly IgG2a antibodies, sustained production of IFN-gamma and a predominantly CD8+ T-cell response. Upon challenge with a virulent S. aureus isolate (Sa042), after 21 days, 55% of the multi-gene vaccinated mice survived infection compared to only 15% of the control groups. Vaccinated mice showed no signs of arthritis when challenged with the less virulent "Newman" strain that caused reactive arthritis in the controls. The results suggest that a multi-gene polyprotein-expressing nucleic acid vaccine alone produces a combined Th1 and Th2 response that can contribute to protection against the complex pathogenesis of S. aureus.


Adhesins, Bacterial/genetics , Aminoacyltransferases/genetics , Bacterial Proteins/genetics , Coagulase/genetics , Cysteine Endopeptidases/genetics , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Bacterial/blood , Antibody Specificity , Female , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Vaccination
...