Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 714052, 2021.
Article in English | MEDLINE | ID: mdl-34603287

ABSTRACT

The T cell immune responses in filarial infections are primarily mediated by CD4+ T cells and type 2-associated cytokines. Emerging evidence indicates that CD8+ T cell responses are important for anti-filarial immunity, however, could be suppressed in co-infections. This review summarizes what we know so far about the activities of CD8+ T cell responses in filarial infections, co-infections, and the associations with the development of filarial pathologies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Susceptibility/immunology , Filariasis/etiology , Filariasis/metabolism , Host-Parasite Interactions/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , Coinfection , Cytokines/metabolism , Disease Models, Animal , Filariasis/prevention & control , Humans , Immunomodulation , Vaccines/immunology
2.
PLoS Pathog ; 15(9): e1008085, 2019 09.
Article in English | MEDLINE | ID: mdl-31568486

ABSTRACT

Human parasitic nematodes are the causative agents of lymphatic filariasis (elephantiasis) and onchocerciasis (river blindness), diseases that are endemic to more than 80 countries and that consistently rank in the top ten for the highest number of years lived with disability. These filarial nematodes have evolved an obligate mutualistic association with an intracellular bacterium, Wolbachia, a symbiont that is essential for the successful development, reproduction, and survival of adult filarial worms. Elimination of the bacteria causes adult worms to die, making Wolbachia a primary target for developing new interventional tools to combat filariases. To further explore Wolbachia as a promising indirect macrofilaricidal drug target, the essential cellular processes that define the symbiotic Wolbachia-host interactions need to be identified. Genomic analyses revealed that while filarial nematodes encode all the enzymes necessary for glycolysis, Wolbachia does not encode the genes for three glycolytic enzymes: hexokinase, 6-phosphofructokinase, and pyruvate kinase. These enzymes are necessary for converting glucose into pyruvate. Wolbachia, however, has the full complement of genes required for gluconeogenesis starting with pyruvate, and for energy metabolism via the tricarboxylic acid cycle. Therefore, we hypothesized that Wolbachia might depend on host glycolysis to maintain a mutualistic association with their parasitic host. We did conditional experiments in vitro that confirmed that glycolysis and its end-product, pyruvate, sustain this symbiotic relationship. Analysis of alternative sources of pyruvate within the worm indicated that the filarial lactate dehydrogenase could also regulate the local intracellular concentration of pyruvate in proximity to Wolbachia and thus help control bacterial growth via molecular interactions with the bacteria. Lastly, we have shown that the parasite's pyruvate kinase, the enzyme that performs the last step in glycolysis, could be a potential novel anti-filarial drug target. Establishing that glycolysis is an essential component of symbiosis in filarial worms could have a broader impact on research focused on other intracellular bacteria-host interactions where the role of glycolysis in supporting intracellular survival of bacteria has been reported.


Subject(s)
Brugia/metabolism , Brugia/microbiology , Pyruvic Acid/metabolism , Wolbachia/metabolism , Animals , Brugia/genetics , Brugia malayi/genetics , Brugia malayi/metabolism , Brugia malayi/microbiology , Brugia pahangi/genetics , Brugia pahangi/metabolism , Brugia pahangi/microbiology , Female , Filariasis/metabolism , Filariasis/microbiology , Filariasis/parasitology , Genes, Helminth , Glycolysis , Host Microbial Interactions , Host-Parasite Interactions , Humans , Male , Symbiosis , Wolbachia/genetics
3.
Dev Cell ; 45(2): 198-211.e3, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29689195

ABSTRACT

Although symbiotic interactions are ubiquitous in the living world, examples of developmental symbioses are still scarce. We show here the crucial role of Wolbachia in the oogenesis of filarial nematodes, a class of parasites of biomedical and veterinary relevance. We applied newly developed techniques to demonstrate the earliest requirements of Wolbachia in the parasite germline preceding the production of faulty embryos in Wolbachia-depleted nematodes. We show that Wolbachia stimulate germline proliferation in a cell-autonomous manner, and not through nucleotide supplementation as previously hypothesized. We also found Wolbachia to maintain the quiescence of a pool of germline stem cells to ensure a constant delivery of about 1,400 eggs per day for many years. The loss of quiescence upon Wolbachia depletion as well as the disorganization of the distal germline suggest that Wolbachia are required to execute the proper germline stem cell developmental program in order to produce viable eggs and embryos.


Subject(s)
Brugia malayi/growth & development , Filariasis/pathology , Germ Cells/cytology , Helminth Proteins/metabolism , Stem Cells/physiology , Symbiosis , Wolbachia/physiology , Animals , Brugia malayi/microbiology , Cell Proliferation , Female , Filariasis/metabolism , Filariasis/parasitology , Germ Cells/microbiology , Germ Cells/physiology , Helminth Proteins/genetics , Male , Stem Cells/cytology , Stem Cells/microbiology
4.
PLoS One ; 12(11): e0188138, 2017.
Article in English | MEDLINE | ID: mdl-29141050

ABSTRACT

Parasitic nematodes have evolved powerful immunomodulatory molecules to enable their survival in immunocompetent hosts by subverting immune responses and minimizing pathological processes. One filarial molecule known to counteract host immune responses by inducing IL-10 and regulatory macrophages in mice is filarial cystatin. During a patent filarial infection monocytes encounter microfilariae in the blood, an event that occurs in asymptomatically infected filariasis patients that are immunologically hyporeactive. The microfilarial larval stage was formerly shown to induce human regulatory monocytes and macrophages. Thus, here we aim was to determine how filarial cystatin of the human pathogenic filaria Brugia malayi (BmCPI-2) contributes to immune hyporesponsiveness in human monocytes and macrophages elicited by microfilaria. For this purpose, filarial cystatin was depleted from microfilarial lysate (Mf). Detecting the immunomodulatory potential of cystatin-depleted Mf revealed that IL-10, but not IL-8 and IL-6 induction in monocytes and macrophages is dependent on the presence of cystatin. In addition, the Mf-induced expression of the regulatory surface markers PD-L1 and PD-L2 in human monocytes, but not in macrophages, is dependent on cystatin. While Mf-treated monocytes result in decreased CD4+ T-cell proliferation in a co-culture assay, stimulation of T-cells with human monocytes treated with cystatin-depleted Mf lead to a restoration of CD4+ T-cell proliferation. Moreover, IL-10 induction by cystatin within Mf was dependent on p38 and ERK in macrophages, but independent of the ERK pathway in monocytes. These findings indicate that filarial nematodes differentially trigger and exploit various signaling pathways to induce immunomodulation in different myeloid cell subsets.


Subject(s)
Brugia malayi/metabolism , Cystatins/pharmacology , Filariasis/metabolism , Macrophages/immunology , Monocytes/immunology , Animals , B7-H1 Antigen/metabolism , Host-Parasite Interactions , Humans , Interleukins/metabolism , Programmed Cell Death 1 Ligand 2 Protein/metabolism
5.
Int J Parasitol ; 47(14): 903-912, 2017 12.
Article in English | MEDLINE | ID: mdl-28743489

ABSTRACT

Onchocerciasis, a neglected tropical disease prevalent in western and central Africa, is a major health problem and has been targeted for elimination. The causative agent for this disease is the human parasite Onchocerca volvulus. Onchocerca ochengi and Litomosoides sigmodontis, infectious agents of cattle and rodents, respectively, serve as model organisms to study filarial nematode infections. Biomarkers to determine infection without the use of painful skin biopsies and microscopic identification of larval worms are needed and their discovery is facilitated by an improved knowledge of parasite-specific metabolites. In addition to proteins and nucleic acids, lipids may be suitable candidates for filarial biomarkers that are currently underexplored. To fill this gap, we present the phospholipid profile of the filarial nematodes O. ochengi, O. volvulus and L. sigmodontis. Direct infusion quadrupole time-of-flight (Q-TOF) mass spectrometry was employed to analyze the composition of phospholipids and their molecular species in the three nematode species. Analysis of the phospholipid profiles of plasma or serum of uninfected and infected hosts showed that nematode-specific phospholipids were below detection limits. However, several phospholipids, in particular ether lipids of phosphatidylethanolamine (PE), were abundant in O. ochengi worms and in bovine nodule fluid, suggesting that these phospholipids might be released from O. ochengi into the host, and could serve as potential biomarkers.


Subject(s)
Filariasis/metabolism , Filarioidea/metabolism , Onchocerca/metabolism , Onchocerciasis/metabolism , Phospholipid Ethers/metabolism , Animals , Biomarkers/metabolism , Cattle , Female , Gerbillinae , Humans , Male , Onchocerca volvulus/metabolism , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
6.
Infect Immun ; 85(1)2017 Jan.
Article in English | MEDLINE | ID: mdl-27799335

ABSTRACT

Filarial parasites cause functional impairment of host dendritic cells (DCs). However, the effects of early infection on individual DC subsets are not known. In this study, we infected BALB/c mice with infective stage 3 larvae of the lymphatic filarial parasite Brugia malayi (Bm-L3) and studied the effect on fluorescence-activated cell sorter (FACS)-sorted DC subsets. While myeloid DCs (mDCs) accumulated by day 3 postinfection (p.i.), lymphoid DCs (LDCs) and CD8+ plasmacytoid DCs (pDCs) peaked at day 7 p.i. in the spleens and mesenteric lymph nodes (mLNs) of infected mice. Increased tumor necrosis factor alpha (TNF-α) but reduced interleukin 12 (IL-12) and Toll-like receptor 4 (TLR4), -6, and -9 and reciprocal secretion of IL-4 and IL-10 were also observed across all DC subsets. Interestingly, Bm-L3 increased the expression of CD80 and CD86 across all DC subsets but decreased that of major histocompatibility complex class II (MHC-II) on mDCs and pDCs, resulting in their impaired antigen uptake and presentation capacities, but maximally attenuated the T-cell proliferation capacity of only mDCs. Furthermore, Bm-L3 increased phosphorylated p38 (p-p38), but not p-ERK, in mDCs and LDCs but downregulated them in pDCs, along with differential modulation of protein tyrosine phosphatases SHP-1, TCPTP, PTEN, and PTP1B across all DC subsets. Taken together, we report hitherto undocumented effects of early Bm-L3 infection on purified host DC subsets that lead to their functional impairment and attenuated host T-cell response.


Subject(s)
Brugia malayi/pathogenicity , Dendritic Cells/pathology , Dendritic Cells/parasitology , Filariasis/pathology , Filariasis/parasitology , Larva/parasitology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/physiology , Dendritic Cells/metabolism , Down-Regulation/physiology , Filariasis/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/physiology , Mice , Mice, Inbred BALB C , PTEN Phosphohydrolase/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
J Infect Dis ; 213(5): 811-5, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26486636

ABSTRACT

Interleukin 19 (IL-19) and interleukin 24 (IL-24) are cytokines that are highly expressed in filarial infections. To study the role of IL-19 and IL-24 in regulating T-cell responses, we examined the frequency of T-helper type 1 (Th1)/Tc1, Th2/Tc2, Th9/Tc9, Th17/Tc17, Th22/Tc22, and Tr1 cells in 26 filariae-infected individuals stimulated with filarial antigen following IL-19 or IL-24 neutralization. IL-19 or IL-24 neutralization resulted in significantly enhanced frequencies of Th1/Tc1 and/or Th17/Tc17 cells and significantly reduced frequencies of Th2/Tc2, Tr1, and/or Th9/Tc9 cells. Thus, we demonstrate that IL-19 and IL-24 are associated with the modulation of T-cell responses in filarial infections.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Filariasis/metabolism , Interleukins/metabolism , Filariasis/immunology , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukins/genetics , T-Lymphocyte Subsets
8.
Arch. Soc. Esp. Oftalmol ; 89(10): 411-413, oct. 2014. ilus
Article in Spanish | IBECS | ID: ibc-128787

ABSTRACT

CASO CLÍNICO: Se presenta un caso de loiasis ocular con una filaria subconjuntival de 5,5 cm de longitud y una microfilaremia grave de una microfilaria/ml, en una paciente previamente asintomática, procedente de Guinea Ecuatorial, con antecedente de hipereosinofilia crónica en estudio. DISCUSSIÓN: La loiasis ocular es una infestación importada y poco frecuente en nuestro medio. No obstante, las parasitaciones crónicas procedentes de inmigrantes de zonas endémicas de África, pueden convertir la loiasis en una enfermedad emergente en nuestro medio


CASE REPORT: We present a case of ocular loiasis with a subconjunctival filaria, 5.5 cm long, and a severe microfilaremia, 1 microfilaria/ml, on a previously asymptomatic woman from Equatorial Guinea, with a past medical history of hypereosinophilia of unknown origin. DISCUSSIÓN: Ocular loiasis is an imported infestation with a very low rate in our country. Nevertheless, chronic infestation in immigrants coming from endemic areas of Africa may increase the rate of this disease in our country (AU)


Subject(s)
Humans , Female , Loiasis/complications , Loiasis/diagnosis , Filariasis/metabolism , Filariasis/parasitology , Eye Infections, Parasitic/diagnosis , Loiasis/chemically induced , Filariasis/diagnosis , Eye Infections, Parasitic/complications
9.
Mol Biochem Parasitol ; 195(1): 54-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25038481

ABSTRACT

Genetic studies undertaken in the model organism Caenorhabditis elegans have demonstrated the importance of neuropeptidergic signalling in nematode physiology. Disruption of this signalling may have deleterious phenotypic consequences, including altered locomotion, feeding behaviour, and reproduction. Neuropeptide G protein-coupled receptors (GPCRs) that transduce many of these signals therefore represent cogent drug targets. Recently published genomic sequencing data for a number of parasitic helminths of medical and veterinary importance has revealed the apparent conservation of a number of neuropeptides, and neuropeptide receptors between parasitic and free-living species, raising the intriguing possibility of developing broad-spectrum anthelmintic therapeutics. Here, we identify and clone a neuropeptide receptor, NPR-4, from the human filarial nematode Brugia malayi and demonstrate its activation in vitro, by FMRFamide-like peptides of the FLP-18 family, and intracellular signalling via Gαi mediated pathways. These data represent the first example of deorphanisation of a neuropeptide GPCR in any parasitic helminth species.


Subject(s)
Brugia malayi/metabolism , Filariasis/metabolism , Filariasis/parasitology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Helminth Proteins/metabolism , Neuropeptides/metabolism , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Animals , Brugia malayi/chemistry , Brugia malayi/genetics , Caenorhabditis elegans , Helminth Proteins/genetics , Humans , Molecular Sequence Data , Neuropeptides/chemistry , Neuropeptides/genetics , Receptors, Neuropeptide/genetics , Sequence Alignment , Signal Transduction
10.
Parasite Immunol ; 36(2): 60-77, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24112106

ABSTRACT

Eosinophil migration as key feature of helminth infection is increased during infection with filarial nematodes. In a mouse model of filariasis, we investigated the role of the eosinophil-attracting chemokine Eotaxin-1 on disease outcome. BALB/c and Eotaxin-1(-/-) mice were infected with the rodent filaria Litomosoides sigmodontis, and parasitic parameters, cellular migration to the site of infection, and cellular responsiveness were investigated. We found increased parasite survival but unaffected eosinophil migration to the site of infection in Eotaxin-1(-/-) mice. Expression of CD80 and CD86 was reduced on eosinophils from Eotaxin-1(-/-) mice after in vitro TLR2 stimulation and exposure to filarial antigen, respectively, suggesting a potential reduced activation state of eosinophils in Eotaxin-1 deficient mice. We further demonstrated that macrophages from Eotaxin-1(-/-) mice produce decreased amounts of IL-6 in vitro, a cytokine found to be associated with parasite containment, suggesting possible mechanisms by which Eotaxin-1 regulates activation of inflammatory cells and thus parasite survival.


Subject(s)
Chemokine CCL11/physiology , Eosinophils/immunology , Filariasis/immunology , Filarioidea/immunology , Macrophages/immunology , Animals , Antigen Presentation , Antigens, Helminth/immunology , Cell Movement , Cells, Cultured , Chemokine CCL11/deficiency , Chemokine CCL11/genetics , Chemokine CCL24/metabolism , Chemokine CCL5/metabolism , Cytokines/metabolism , Eosinophils/physiology , Epithelial Cells/metabolism , Female , Filariasis/metabolism , Filariasis/parasitology , Filarioidea/growth & development , Interleukin-6/metabolism , Macrophage Activation , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Microfilariae/physiology , Parasite Load , Pleural Cavity/immunology , Pleural Cavity/parasitology , Spleen/immunology
11.
J Immunol ; 188(9): 4188-99, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22461700

ABSTRACT

Basophils play a key role in the development and effector phases of type 2 immune responses in both allergic diseases and helminth infections. This study shows that basophils become less responsive to IgE-mediated stimulation when mice are chronically infected with Litomosoides sigmodontis, a filarial nematode, and Schistosoma mansoni, a blood fluke. Although excretory/secretory products from microfilariae of L. sigmodontis suppressed basophils in vitro, transfer of microfilariae into mice did not result in basophil suppression. Rather, reduced basophil responsiveness, which required the presence of live helminths, was found to be dependent on host IL-10 and was accompanied by decreases in key IgE signaling molecules known to be downregulated by IL-10. Given the importance of basophils in the development of type 2 immune responses, these findings help explain the mechanism by which helminths protect against allergy and may have broad implications for understanding how helminth infections alter other disease states in people.


Subject(s)
Basophils/immunology , Filariasis/immunology , Filarioidea/immunology , Interleukin-10/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Basophils/metabolism , Chronic Disease , Down-Regulation/genetics , Down-Regulation/immunology , Female , Filariasis/genetics , Filariasis/metabolism , Filarioidea/metabolism , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/immunology , Th2 Cells/metabolism
12.
J Immunol ; 188(2): 559-68, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22174447

ABSTRACT

Leading hypotheses to explain helminth-mediated protection against autoimmunity postulate that type 2 or regulatory immune responses induced by helminth infections in the host limit pathogenic Th1-driven autoimmune responses. We tested these hypotheses by investigating whether infection with the filarial nematode Litomosoides sigmodontis prevents diabetes onset in IL-4-deficient NOD mice and whether depletion or absence of regulatory T cells, IL-10, or TGF-ß alters helminth-mediated protection. In contrast to IL-4-competent NOD mice, IL-4-deficient NOD mice failed to develop a type 2 shift in either cytokine or Ab production during L. sigmodontis infection. Despite the absence of a type 2 immune shift, infection of IL-4-deficient NOD mice with L. sigmodontis prevented diabetes onset in all mice studied. Infections in immunocompetent and IL-4-deficient NOD mice were accompanied by increases in CD4(+)CD25(+)Foxp3(+) regulatory T cell frequencies and numbers, respectively, and helminth infection increased the proliferation of CD4(+)Foxp3(+) cells. However, depletion of CD25(+) cells in NOD mice or Foxp3(+) T cells from splenocytes transferred into NOD.scid mice did not decrease helminth-mediated protection against diabetes onset. Continuous depletion of the anti-inflammatory cytokine TGF-ß, but not blockade of IL-10 signaling, prevented the beneficial effect of helminth infection on diabetes. Changes in Th17 responses did not seem to play an important role in helminth-mediated protection against autoimmunity, because helminth infection was not associated with a decreased Th17 immune response. This study demonstrates that L. sigmodontis-mediated protection against diabetes in NOD mice is not dependent on the induction of a type 2 immune shift but does require TGF-ß.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/parasitology , Filariasis/immunology , Filariasis/parasitology , Filarioidea/immunology , Th1 Cells/immunology , Transforming Growth Factor beta/biosynthesis , Animals , Diabetes Mellitus, Type 1/metabolism , Female , Filariasis/metabolism , Interleukin-10/biosynthesis , Interleukin-10/physiology , Interleukin-4/deficiency , Interleukin-4/genetics , Mice , Mice, 129 Strain , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/parasitology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/metabolism , Th1 Cells/parasitology , Transforming Growth Factor beta/physiology
13.
Int J Biol Sci ; 7(9): 1371-81, 2011.
Article in English | MEDLINE | ID: mdl-22110388

ABSTRACT

Helminth parasites are a diverse group of multicellular organisms. Despite their heterogeneity, helminths share many common characteristics, such as the modulation of the immune system of their hosts towards a permissive state that favors their development. They induce strong Th2-like responses with high levels of IL-4, IL-5 and IL-13 cytokines, and decreased production of proinflammatory cytokines such as IFN-γ. IL-4, IFN-γ and other cytokines bind with their specific cytokine receptors to trigger an immediate signaling pathway in which different tyrosine kinases (e.g. Janus kinases) are involved. Furthermore, a seven-member family of transcription factors named Signal Transducers and Activators of Transcription (STAT) that initiate the transcriptional activation of different genes are also involved and regulate downstream the JAK/STAT signaling pathway. However, how helminths avoid and modulate immune responses remains unclear; moreover, information concerning STAT-mediated immune regulation during helminth infections is scarce. Here, we review the research on mice deficient in STAT molecules, highlighting the importance of the JAK/STAT signaling pathway in regulating susceptibility and/or resistance in these infections.


Subject(s)
STAT Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Cytokines/metabolism , Filariasis/metabolism , Helminthiasis/metabolism , Helminths/metabolism , Helminths/pathogenicity , Receptors, Cytokine/metabolism , STAT Transcription Factors/genetics , Schistosomiasis/metabolism , Signal Transduction/genetics , Taenia/pathogenicity
14.
J Leukoc Biol ; 90(5): 845-54, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21628332

ABSTRACT

Macrophages become activated by their environment and develop polarized functions: classically activated (M1) macrophages eliminate pathogens but can cause tissue injury, whereas alternatively activated (M2) macrophages promote healing and repair. Mechanisms directing polarized activation, especially in vivo, are not understood completely, and here, we examined the role of SOCS proteins. M2 macrophages activated in vitro or elicited by implanting mice i.p. with the parasitic nematode Brugia malayi display a selective and IL-4-dependent up-regulation of SOCS1 but not SOCS3. Using siRNA-targeted knockdown in BMDM, we reveal that the enhanced SOCS1 is crucial for IL-4-induced M2 characteristics, including a high arginase I:iNOS activity ratio, suppression of T cell proliferation, attenuated responses to IFN-γ/LPS, and curtailed SOCS3 expression. Importantly, SOCS1 was essential in sustaining the enhanced PI3K activity that drives M2 activation, defining a new regulatory mechanism by which SOCS1 controls M2 polarization. By contrast, for M1 macrophages, SOCS1 was not only an important regulator of proinflammatory mediators (IL-6, IL-12, MHC class II, NO), but critically, for M1, we show that SOCS1 also restricted IL-10 secretion and arginase I activity, which otherwise would limit the efficiency of M1 macrophage proinflammatory responses. Together, our results uncover SOCS1, not only as a feedback inhibitor of inflammation but also as a critical molecular switch that tunes key signaling pathways to effectively program different sides of the macrophage balance.


Subject(s)
Macrophage Activation , Macrophages , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Arginase/metabolism , Brugia malayi/immunology , Filariasis/immunology , Filariasis/metabolism , Inflammation , Interferon-gamma/pharmacology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Up-Regulation , Wound Healing
15.
Mol Biochem Parasitol ; 176(2): 80-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21241743

ABSTRACT

Juv-p120 is an excretory-secretory 160 kDa glycoprotein of juvenile female Litomosoides sigmodontis and exhibits features typical for mucins. 50% of its molecular mass is attributed to posttranslational modifications with the unusual substituent dimethylaminoethanol (DMAE). By that Juv-p120 corresponds to the surface proteins of the microfilarial sheath, Shp3 and Shp3a. The secreted protein consists of 697 amino acids, organized in two different domains of repeat elements separated by a stretch of polar residues. The N-terminal domain shows fourteen P/S/T/F-rich repeat elements highly modified with phospho-DMAE substituted O-glycans confering a negative charge to the protein. The C-terminal domain is extremely rich in glutamine (35%) and leucine (25%) in less organized repeats and may play a role in oligomerization of Juv-p120 monomers. A protein family with a similar Q/L-rich region and conserved core promoter region was identified in Brugia malayi by homology screening and in Wuchereria bancrofti and Loa loa by database similarity search. One of the Q/L-rich proteins in each genus has an extended S/T-rich region and due to this feature is supposed to be a putative Juv-p120 ortholog. The corresponding modification of Juv-p120 and the microfilarial sheath surface antigens Shp3/3a explains the appearance of anti-sheath antibodies before the release of microfilariae. The function of Juv-p120 is unknown.


Subject(s)
Antigens, Helminth/genetics , Deanol/metabolism , Filarioidea/chemistry , Membrane Proteins/genetics , Microfilariae/chemistry , Amino Acid Motifs , Amino Acid Sequence , Amino Acids/genetics , Amino Acids/metabolism , Animals , Antibodies, Helminth/immunology , Antigens, Helminth/immunology , Antigens, Helminth/metabolism , Brugia malayi , Deanol/chemistry , Female , Filariasis/genetics , Filariasis/immunology , Filariasis/metabolism , Filarioidea/genetics , Filarioidea/immunology , Filarioidea/metabolism , Loa , Membrane Proteins/immunology , Membrane Proteins/metabolism , Microfilariae/genetics , Microfilariae/immunology , Microfilariae/metabolism , Molecular Sequence Data , Molecular Weight , Murinae , Protein Processing, Post-Translational , Sequence Homology , Wuchereria bancrofti
16.
Mol Biochem Parasitol ; 176(2): 90-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21185874

ABSTRACT

Filariasis, caused by thread-like nematode worms, affects millions of individuals throughout the tropics and is a major cause of acute and chronic morbidity. Filarial nematodes effectively evade host immunological responses and are long lived within their hosts. Recently an emphasis has been placed on enzymatic and non-enzymatic anti-oxidant systems which counteract the generation of reactive oxygen species (ROS) by macrophages and granulocytes, a first line of defense against parasites. We have characterized an anti-oxidant pathway in the filarial parasite Brugia malayi related to the evolutionarily conserved human mitogen-activated p38 protein kinase and the Caenorhabditis elegans PMK-1 protein kinase stress pathways. We have expressed a recombinant p38/PMK-1 ortholog from B. malayi (Bm-MPK1) and have successfully activated the kinase with mammalian upstream kinases. In addition, we have demonstrated inhibition of Bm-MPK1 activity using a panel of known p38 inhibitors. Using the potent and highly selective allosteric p38 inhibitor, BIRB796, we have implicated Bm-MPK1 in a pathway which offers B. malayi protection from the effects of ROS. Our results, for the first time, describe a stress-activated protein kinase pathway within the filarial parasite B. malayi which plays a role in protecting the parasite from ROS. Inhibition of this pathway may have therapeutic benefit in treating filariasis by increasing the sensitivity of filarial parasites to ROS and other reactive intermediates.


Subject(s)
Brugia malayi/metabolism , Helminth Proteins/metabolism , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Sequence , Animals , Brugia malayi/drug effects , Brugia malayi/genetics , Caenorhabditis elegans , Female , Filariasis/drug therapy , Filariasis/genetics , Filariasis/metabolism , Gene Expression , HEK293 Cells , Helminth Proteins/genetics , Humans , Molecular Sequence Data , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , p38 Mitogen-Activated Protein Kinases/genetics
17.
J Biol Chem ; 284(33): 22364-22378, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19458089

ABSTRACT

Wolbachia endosymbiotic bacteria have been implicated in the inflammatory pathogenesis of filariasis. Inflammation induced by Brugia malayi female worm extract (BMFE) is dependent on Toll-like receptors 2 and 6 (TLR2/6) with only a partial requirement for TLR1. Removal of Wolbachia, lipids, or proteins eliminates all inflammatory activity. Wolbachia bacteria contain the lipoprotein biosynthesis genes Ltg and LspA but not Lnt, suggesting Wolbachia proteins cannot be triacylated, accounting for recognition by TLR2/6. Lipoprotein databases revealed 3-11 potential lipoproteins from Wolbachia. Peptidoglycan-associated lipoprotein (PAL) and Type IV secretion system-VirB6 were consistently predicted, and B. malayi Wolbachia PAL (wBmPAL) was selected for functional characterization. Diacylated 20-mer peptides of wBmPAL (Diacyl Wolbachia lipopeptide (Diacyl WoLP)) showed a near identical TLR2/6 and TLR2/1 usage compared with BMFE and bound directly to TLR2. Diacyl WoLP induced systemic tumor necrosis factor-alpha and neutrophil-mediated keratitis in mice. Diacyl WoLP activated monocytes induce up-regulation of gp38 on human lymphatic endothelial cells and induced dendritic cell maturation and activation. Dendritic cells primed with BMFE generated a non-polarized Th1/Th2 CD4+ T cell profile, whereas priming with Wolbachia depleted extracts (following tetracycline treatment; BMFEtet) polarized to a Th2 profile that could be reversed by reconstitution with Diacyl WoLP. BMFE generated IgG1 and IgG2c antibody responses, whereas BMFEtet or inoculation of TLR2 or MyD88-/- mice produced defective IgG2c responses. Thus, in addition to innate inflammatory activation, Wolbachia lipoproteins drive interferon-gamma-dependent CD4+ T cell polarization and antibody switching.


Subject(s)
Brugia/metabolism , Filariasis/metabolism , Immune System , Lipoproteins/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/metabolism , Wolbachia/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Transgenic , Models, Biological
18.
J Immunol ; 181(9): 6456-66, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941236

ABSTRACT

Many helminths, including Brugia malayi, are able to establish long-lived infections in immunocompetent hosts. Growing evidence suggests that the immune system's failure to eliminate parasites is at least partially due to the effects of regulatory T cells (Tregs). To test whether parasites may directly stimulate host regulatory activity, we infected mice with two key stages of B. malayi. Both mosquito-borne infective larvae and mature adults i.p. introduced were found to preferentially expand the proportion of CD25(+)Foxp3(+) cells within the CD4(+) T cell population. The induction of Foxp3 was accompanied by raised CD25, CD103, and CTLA-4 expression, and was shown to be an active process, which accompanied the introduction of live, but not dead parasites. CTLA-4 expression was also markedly higher on Foxp3(-) cells, suggesting anergized effector populations. Peritoneal lavage CD4(+)CD25(+) cells from infected mice showed similar suppressive activity in vitro to normal splenic "natural" Tregs. Both B. malayi larvae and adults were also able to induce Foxp3 expression in adoptively transferred DO11.10 T cells, demonstrating that filarial infection can influence the development of T cells specific to a third party Ag. In addition, we showed that induction was intact in IL-4R-deficient animals, in the absence of a Th2 or alternatively activated macrophage response. We conclude that filarial infections significantly skew the balance of the host immune system toward Treg expansion and activation, in a manner dependent on live parasites but independent of a concomitant Th2 response.


Subject(s)
Brugia malayi/immunology , Forkhead Transcription Factors/biosynthesis , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/parasitology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Movement/immunology , Cells, Cultured , Filariasis/immunology , Filariasis/metabolism , Filariasis/pathology , Gerbillinae , Humans , Macrophage Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-4/deficiency , Receptors, Interleukin-4/genetics , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/transplantation
19.
Biopharm Drug Dispos ; 28(8): 431-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17847063

ABSTRACT

This study compares plasma disposition kinetics of ivermectin and moxidectin after oral administration to beagle dogs experimentally infected with the filarial parasite, Brugia pahangi. Sixteen dogs were selected and randomly allocated into two groups of eight dogs each. Animals in each group received either ivermectin or moxidectin by oral route at a dose of 250 microg/kg. Blood samples were collected from 0.5 h up to 56 days post-treatment and the plasma was analysed by high performance liquid chromatography (HPLC). The obtained data were analysed by compartmental and non-compartmental pharmacokinetic techniques. Peak plasma concentrations (C(max)) of 234.0 +/- 64.3 ng/ml (mean +/- SD) were obtained for moxidectin and 132.6 +/- 43.0 ng/ml for ivermectin. The terminal elimination half-life was significantly (p<0.01) longer in the moxidectin treated group (621.3 +/- 149.3 h) than for ivermectin treated group (80.3 +/- 29.8 h). A significantly (p< 0.01) larger V(ss)/F was obtained for moxidectin (19.21 +/- 3.61 l/kg) compared with ivermectin (5.35 +/- 1.29 l/kg). The mean estimates of CL/F of moxidectin and ivermectin were 0.0220 +/- 0.00381 and 0.0498 +/- 0.0179 l/h/kg, respectively. The comparative plasma disposition kinetics of ivermectin and moxidectin in dogs is reported for the first time.


Subject(s)
Filaricides/pharmacokinetics , Ivermectin/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Brugia pahangi , Chromatography, High Pressure Liquid , Data Interpretation, Statistical , Dogs , Female , Filariasis/metabolism , Half-Life , Indicators and Reagents , Macrolides/pharmacokinetics , Male , Spectrometry, Fluorescence
20.
J Immunol ; 179(4): 2445-56, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17675506

ABSTRACT

NK cells are an important source of early cytokine production in a variety of intracellular viral, bacterial, and protozoan infections; however, the role of NK cells in extracellular parasitic infections such as filarial infections is not well-defined. To investigate the role of NK cells in filarial infections, we have used an in vitro model system of culturing live infective-stage larvae (L3) or live microfilariae (Mf) of Brugia malayi, a causative agent of human lymphatic filariasis, with PBMC of normal individuals. We found that NK cells undergo early cell activation and produce IFN-gamma and TNF-alpha within 24 h after stimulation with both live L3 and Mf. Interestingly, NK cells also express IL-4 and IL-5 at this time point in response to live Mf but not L3. This is accompanied by significant alterations in NK cell expression of costimulatory molecules and natural cytotoxicity receptors. This activation is dependent on the presence of monocytes in the culture, IL-12, and direct contact with live parasites. The early activation event is subsequently followed by apoptosis of NK cells involving a caspase-dependent mechanism in response to live L3 but not live Mf. Thus, the NK cell-parasite interaction is complex, with filarial parasites inducing NK cell activation and cytokine secretion and finally NK cell apoptosis, which may provide an additional mechanism of down-regulating the host immune response.


Subject(s)
Apoptosis/immunology , Brugia malayi/immunology , Cytokines/immunology , Filariasis/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Wuchereria bancrofti/immunology , Animals , Cytokines/metabolism , Down-Regulation/immunology , Filariasis/metabolism , Filariasis/parasitology , Host-Parasite Interactions/immunology , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/parasitology , Larva/immunology , Models, Immunological , Th1 Cells/metabolism , Th1 Cells/parasitology , Th2 Cells/metabolism , Th2 Cells/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...