Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Sci Transl Med ; 15(719): eadj0740, 2023 10 25.
Article En | MEDLINE | ID: mdl-37878675

Recurrent respiratory papillomatosis (RRP) is a rare, debilitating neoplastic disorder caused by chronic infection with human papillomavirus (HPV) type 6 or 11 and characterized by growth of papillomas in the upper aerodigestive tract. There is no approved medical therapy, and patients require repeated debulking procedures to maintain voice and airway function. PRGN-2012 is a gorilla adenovirus immune-therapeutic capable of enhancing HPV 6/11-specific T cell immunity. This first-in-human, phase 1 study (NCT04724980) of adjuvant PRGN-2012 treatment in adult patients with severe, aggressive RRP demonstrates the overall safety and clinically meaningful benefit observed with PRGN-2012, with a 50% complete response rate in patients treated at the highest dose. Responders demonstrate greater expansion of peripheral HPV-specific T cells compared with nonresponders. Additional correlative studies identify an association between reduced baseline papilloma HPV gene expression, greater interferon responses and expression of CXCL9 and CXCL10, and greater papilloma T cell infiltration in responders. Conversely, nonresponders were characterized by greater HPV and CXCL8 gene expression, increased neutrophilic cell infiltration, and reduced T cell papilloma infiltration. These results suggest that papilloma HPV gene expression may regulate interferon signaling and chemokine expression profiles within the tumor microenvironment that cooperate to govern clinical response to therapeutic HPV vaccination in patients with respiratory papillomatosis.


Papilloma , Papillomavirus Infections , Respiratory Tract Infections , Adult , Humans , Papillomavirus Infections/therapy , Papillomavirus Infections/pathology , Tumor Microenvironment , Respiratory Tract Infections/therapy , Interferons , Papilloma/therapy , Papilloma/pathology , Vaccination
2.
Int J Mol Sci ; 24(13)2023 Jun 24.
Article En | MEDLINE | ID: mdl-37445745

This study investigated the role of IL-35 in systemic sclerosis (SSc) patients, focusing on CD4+ T cell response and immunomodulatory cytokine production. By comparing the cytokine levels in healthy donors (HD) and SSc patients using ELISAs, we found a significantly lower plasma IL-35 concentration in the SSc patients (52.1 ± 5.6 vs. 143 ± 11.1, p < 0.001). Notably, the IL-35 levels showed a negative correlation with TGF-ß (p < 0.001) and IL-17 (p = 0.04). Assessing the IL-35R expression across cell types in the SSc patients and HDs via flow cytometry, we found higher levels on monocytes (40.7 + 5.7 vs. 20.3 ± 1.9, p < 0.001) and lower levels on CD8+ T cells (61.8 ± 9.2 vs. 83.4 ± 0.8, p < 0.05) in the SSc patients. The addition of recombinant IL-35 to stimulated peripheral blood mononuclear cells reduced the IL-17+CD4+ T cell percentage (9.0 ± 1.5 vs. 4.8 ± 0.7, p < 0.05) and increased the IL-35+CD4+ T percentage (4.1 ± 2.3 vs. 10.2 ± 0.8, p < 0.001). In a Treg:Tresponder cell Sco-culture assay with HD and SSc samples, rIL35 decreased the cell proliferation and levels of IL-17A (178.2 ± 30.5 pg/mL vs. 37.4 ± 6.4 pg/mL, p < 0.001) and TGF-ß (4194 ± 777 pg/mL vs. 2413 ± 608 pg/mL, p < 0.01). Furthermore, we observed a positive correlation between the modified Rodnan skin score (mRSS) and TGF-ß (p < 0.001), while there was a negative correlation between mRSS and IL-35 (p = 0.004). Interestingly, higher levels of plasmatic IL-35 were detected in individuals with limited disease compared to those with diffuse disease (60.1 ± 8.0 vs. 832.3 ± 4.1, p < 0.05). These findings suggest that IL-35 exhibits anti-inflammatory properties in SSc and it may serve as a marker for disease severity and a therapeutic target.


Interleukin-17 , Scleroderma, Systemic , Humans , Interleukin-17/metabolism , Leukocytes, Mononuclear/metabolism , Scleroderma, Systemic/metabolism , Cytokines/metabolism , Transforming Growth Factor beta
3.
Immunology ; 168(4): 610-621, 2023 04.
Article En | MEDLINE | ID: mdl-36273280

Pulmonary fibrosis involves various types of immune cells and soluble mediators, including TGF-ß and IL-35, a recently identified heterodimeric cytokine that belongs to the IL-12 cytokine family. However, the effect of regulatory IL-35 may play an important role in fibrotic diseases. The aim of this paper is to explore the immunoregulatory role of IL-35 in the development of fibrosis in interstitial lung disease (ILD). To gain a better understanding of this issue, the concentrations of IL-35 and different profibrotic cytokines in fibrotic (F-ILD) and non-fibrotic (NF-ILD) patients by ELISA were compared to that of intracellular IL-35 and IL-17 on CD4+ T cells stimulated in the presence of BAL or with different ratios of recombinant IL-35 (rIL-35) and TGF-ß (rTGF-ß), which were evaluated by flow cytometry. We observed that BAL concentration of IL-35 was lower in F patients (p < 0.001) and was negatively correlated with concentrations of TGF-ß (p < 0.001) and IL-17 (p < 0.001). In supplemented cell cultures, BAL from NF but not F patients enhanced the percentage of IL-35 + CD4+ T (p < 0.001) cells and decreased the percentage of IL-17 + CD4+ T cells (p < 0.001). The percentage of IL-35 + CD4+ T cells correlated positively with BAL concentration of IL-35 (p = 0.02), but correlated negatively with BAL concentrations of IL-17 (p = 0.007) and TGF-ß (p = 0.01). After adjusting the concentrations of recombinant cytokines to establish a TGF-ß: IL-35 ratio of 1:4, an enhanced percentage of IL-35 + CD4+ T cells (p < 0.001) but a decreased percentage of IL-17 + CD4+ T cells (p < 0.001) was observed. After adding recombinant IL-35 to the BAL from F patients until a 1:4 ratio of TGF-ß: IL-35 was reached, a significantly increased percentage of IL-35 + CD4+ T cells (p < 0.001) and a decreased percentage of IL-17 + CD4+ T cells (p = 0.003) was found. These results suggest that IL-35 may induce an anti-fibrotic response, regulating the effect of TGF-ß and the inflammatory response on CD4+ T cells. In addition, the TGF-ß: IL-35 ratio in BAL has been shown to be a potential biomarker to predict the outcome of F patients with ILD.


Lung Diseases, Interstitial , Pulmonary Fibrosis , Humans , Interleukin-17 , Cytokines/analysis , Transforming Growth Factor beta , Bronchoalveolar Lavage Fluid
4.
Am J Trop Med Hyg ; 2022 Feb 28.
Article En | MEDLINE | ID: mdl-35226872

Monocyte dysfunction in helminth infection is one of the mechanisms proposed to explain the diminished parasite antigen-specific T cell responses seen with patent filarial infection. In fact, monocytes from filariae-infected individuals demonstrate internalized filarial antigens and, as a consequence, express inhibitory surface molecules and have diminished cytokine production. To investigate the mechanisms underlying monocyte dysfunction in filarial infections, purified human monocytes were exposed to live microfilariae (mf) of Brugia malayi, and the mRNA and protein expression of important inhibitory and/or autophagy-related molecules were assessed. Our data indicate that mf-induced autophagy in human monocytes shown by the formation of autophagic vesicles, by the upregulation in the mRNA expression of autophagy-related genes BCN1, LC3B, ATG5, ATG7 (P < 0.05), and by increase in the levels of LC3B protein. Furthermore, this mf-induced autophagy increased the levels of monocyte CD206 expression. In addition, mf significantly induced the frequency of interferon (IFN)-γ+ human monocytes and at the same time induced the mRNA expression of indoleamine 2,3-dioxygenase (IDO) through an IFN-γ-dependent mechanism; significantly enhanced tryptophan degradation (an indicator of IDO activity; P < 0.005). Interestingly, this autophagy induction by mf in monocytes was IFN-γ-dependent but IDO-independent as was reversed by anti-IFN-γ but not by an IDO inhibitor. Our data collectively suggest that mf of Brugia malayi regulate the function of monocytes by induction of IDO and IFN-γ, induce autophagy through an IFN-γ-dependent mechanism, and increase M2 phenotype through induction of autophagy; all acting in concert to drive monocyte dysfunction.

5.
PLoS Negl Trop Dis ; 15(6): e0009448, 2021 06.
Article En | MEDLINE | ID: mdl-34106920

BACKGROUND: In Mali, cutaneous leishmaniasis (CL) and filariasis are co-endemic. Previous studies in animal models of infection have shown that sand fly saliva enhance infectivity of Leishmania parasites in naïve hosts while saliva-specific adaptive immune responses may protect against cutaneous and visceral leishmaniasis. In contrast, the human immune response to Phlebotomus duboscqi (Pd) saliva, the principal sand fly vector in Mali, was found to be dichotomously polarized with some individuals having a Th1-dominated response and others having a Th2-biased response. We hypothesized that co-infection with filarial parasites may be an underlying factor that modulates the immune response to Pd saliva in endemic regions. METHODOLOGY/PRINCIPAL FINDINGS: To understand which cell types may be responsible for polarizing human responses to sand fly saliva, we investigated the effect of salivary glands (SG) of Pd on human monocytes. To this end, elutriated monocytes were cultured in vitro, alone, or with SG, microfilariae antigen (MF ag) of Brugia malayi, or LPS, a positive control. The mRNA expression of genes involved in inflammatory or regulatory responses was then measured as were cytokines and chemokines associated with these responses. Monocytes of individuals who were not exposed to sand fly bites (mainly North American controls) significantly upregulated the production of IL-6 and CCL4; cytokines that enhance leishmania parasite establishment, in response to SG from Pd or other vector species. This selective inflammatory response was lost in individuals that were exposed to sand fly bites which was not changed by co-infection with filarial parasites. Furthermore, infection with filarial parasites resulted in upregulation of CCL22, a type-2 associated chemokine, both at the mRNA levels and by its observed effect on the frequency of recruited monocytes. CONCLUSIONS/SIGNIFICANCE: Together, our data suggest that SG or recombinant salivary proteins from Pd alter human monocyte function by upregulating selective inflammatory cytokines.


Brugia malayi/immunology , Insect Proteins/immunology , Monocytes/parasitology , Phlebotomus/immunology , Saliva/immunology , Adaptive Immunity , Animals , Cells, Cultured , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Coinfection , Endemic Diseases , Filariasis/complications , Filariasis/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Immunity, Cellular , Leishmaniasis, Cutaneous/complications , Leishmaniasis, Cutaneous/immunology , Lipopolysaccharides/toxicity , Mali , Monocytes/physiology , RNA, Messenger , Recombinant Proteins , Salivary Glands , T-Lymphocytes, Helper-Inducer
6.
PLoS Negl Trop Dis ; 15(1): e0008884, 2021 01.
Article En | MEDLINE | ID: mdl-33411714

We have previously shown that the microfilarial (mf) stage of Brugia malayi can inhibit the mammalian target of rapamycin (mTOR; a conserved serine/threonine kinase critical for immune regulation and cellular growth) in human dendritic cells (DC) and we have proposed that this mTOR inhibition is associated with the DC dysfunction seen in filarial infections. Extracellular vesicles (EVs) contain many proteins and nucleic acids including microRNAs (miRNAs) that might affect a variety of intracellular pathways. Thus, EVs secreted from mf may elucidate the mechanism by which the parasite is able to modulate the host immune response during infection. EVs, purified from mf of Brugia malayi and confirmed by size through nanoparticle tracking analysis, were assessed by miRNA microarrays (accession number GSE157226) and shown to be enriched (>2-fold, p-value<0.05, FDR = 0.05) for miR100, miR71, miR34, and miR7. The microarray analysis compared mf-derived EVs and mf supernatant. After confirming their presence in EVs using qPCR for these miRNA targets, web-based target predictions (using MIRPathv3, TarBAse and MicroT-CD) predicted that miR100 targeted mTOR and its downstream regulatory protein 4E-BP1. Our previous data with live parasites demonstrated that mf downregulate the phosphorylation of mTOR and its downstream effectors. Additionally, our proteomic analysis of the mf-derived EVs revealed the presence of proteins commonly found in these vesicles (data are available via ProteomeXchange with identifier PXD021844). We confirmed internalization of mf-derived EVs by human DCs and monocytes using confocal microscopy and flow cytometry, and further demonstrated through flow cytometry, that mf-derived EVs downregulate the phosphorylation of mTOR in human monocytes (THP-1 cells) to the same degree that rapamycin (a known mTOR inhibitor) does. Our data collectively suggest that mf release EVs that interact with host cells, such as DC, to modulate host responses.


Brugia malayi/metabolism , Down-Regulation , Extracellular Vesicles/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brugia malayi/immunology , Cell Cycle Proteins/metabolism , Dendritic Cells/immunology , Filariasis/immunology , Humans , MicroRNAs/metabolism , Microfilariae/immunology , Monocytes/metabolism , Phosphorylation , Proteomics , THP-1 Cells , TOR Serine-Threonine Kinases/genetics
7.
PLoS Negl Trop Dis ; 12(4): e0006404, 2018 04.
Article En | MEDLINE | ID: mdl-29668679

A number of features at the host-parasite interface are reminiscent of those that are also observed at the host-tumor interface. Both cancer cells and parasites establish a tissue microenvironment that allows for immune evasion and may reflect functional alterations of various innate cells. Here, we investigated how the phenotype and function of human monocytes is altered by exposure to cancer cell lines and if these functional and phenotypic alterations parallel those induced by exposure to helminth parasites. Thus, human monocytes were exposed to three different cancer cell lines (breast, ovarian, or glioblastoma) or to live microfilariae (mf) of Brugia malayi-a causative agent of lymphatic filariasis. After 2 days of co-culture, monocytes exposed to cancer cell lines showed markedly upregulated expression of M1-associated (TNF-α, IL-1ß), M2-associated (CCL13, CD206), Mreg-associated (IL-10, TGF-ß), and angiogenesis associated (MMP9, VEGF) genes. Similar to cancer cell lines, but less dramatically, mf altered the mRNA expression of IL-1ß, CCL13, TGM2 and MMP9. When surface expression of the inhibitory ligands PDL1 and PDL2 was assessed, monocytes exposed to both cancer cell lines and to live mf significantly upregulated PDL1 and PDL2 expression. In contrast to exposure to mf, exposure to cancer cell lines increased the phagocytic ability of monocytes and reduced their ability to induce T cell proliferation and to expand Granzyme A+ CD8+ T cells. Our data suggest that despite the fact that helminth parasites and cancer cell lines are extraordinarily disparate, they share the ability to alter the phenotype of human monocytes.


Brugia malayi/immunology , Filariasis/immunology , Immune Evasion , Monocytes/immunology , Monocytes/parasitology , Neoplasms/immunology , Animals , Brugia malayi/genetics , Brugia malayi/physiology , Cell Line, Tumor , Filariasis/parasitology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lymphocyte Activation , Phagocytosis , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
8.
Parasit Vectors ; 11(1): 18, 2018 01 08.
Article En | MEDLINE | ID: mdl-29310700

BACKGROUND: Filariasis and leishmaniasis are two neglected tropical diseases in Mali. Due to distribution and associated clinical features, both diseases are of concern to public health. The goal of this study was to determine the prevalence of co-infection with filarial (Wuchereria bancrofti and Mansonella perstans) and Leishmania major parasites in two ecologically distinct areas of Mali, the Kolokani district (villages of Tieneguebougou and Bougoudiana) in North Sudan Savanna area, and the district of Kolondieba (village of Boundioba) in the South Sudan Savanna area. METHODS: The prevalence of co-infection (filarial and Leishmania) was measured based on (i) Mansonella perstans microfilaremia count and/or filariasis immunochromatographic test (ICT) for Wuchereria bancrofti-specific circulating antigen, and (ii) the prevalence of delayed type hypersensitivity (DTH) responses to Leishmania measured by leishmanin skin test (LST). RESULTS: In this study, a total of 930 volunteers between the age of 18 and 65 were included from the two endemic areas of Kolokani and Kolondieba. In general, in both areas, filarial infection was more prevalent than Leishmania infection with an overall prevalence of 15.27% (142/930) including 8.7% (81/930) for Mansonella perstans and 8% (74/930) for Wuchereria bancrofti-specific circulating antigen. The prevalence of Leishmania major infection was 7.7% (72/930) and was significantly higher in Tieneguebougou and Bougoudiana (15.05%; 64/425) than in Boundioba (2.04%; 8/505) (χ2 = 58.66, P < 0.0001). Among the filarial infected population, nearly 10% (14/142) were also positive for Leishmania with an overall prevalence of co-infection of 1.50% (14/930) varying from 2.82% (12/425) in Tieneguebougou and Bougoudiana to 0.39% (2/505) in Boundioba (P = 0.0048). CONCLUSION: This study established the existence of co-endemicity of filarial and Leishmania infections in specific regions of Mali. Since both filarial and Leishmania infections are vector-borne with mosquitoes and sand flies as respective vectors, an integrated vector control approach should be considered in co-endemic areas. The effect of potential interaction between filarial and Leishmania parasites on the disease outcomes may be further studied.


Coinfection/epidemiology , Endemic Diseases , Filariasis/epidemiology , Leishmaniasis/epidemiology , Adult , Animals , Chromatography, Affinity , Cross-Sectional Studies , Female , Filariasis/complications , Healthy Volunteers , Humans , Leishmaniasis/complications , Male , Mali/epidemiology , Microscopy , Middle Aged , Prevalence , Skin Tests , Sudan/epidemiology , Young Adult
9.
Infect Immun ; 84(9): 2463-72, 2016 09.
Article En | MEDLINE | ID: mdl-27297394

Immune modulation is a hallmark of patent filarial infection, including suppression of antigen-presenting cell function and downmodulation of filarial antigen-specific T cell responses. The mammalian target of rapamycin (mTOR) signaling pathway has been implicated in immune regulation, not only by suppressing T cell responses but also by regulating autophagy (through mTOR sensing amino acid availability). Global proteomic analysis (liquid chromatography-tandem mass spectrometry) of microfilaria (mf)-exposed monocyte-derived dendritic cells (DC) indicated that multiple components of the mTOR signaling pathway, including mTOR, eIF4A, and eIF4E, are downregulated by mf, suggesting that mf target this pathway for immune modulation in DC. Utilizing Western blot analysis, we demonstrate that similar to rapamycin (a known mTOR inhibitor), mf downregulate the phosphorylation of mTOR and its regulatory proteins, p70S6K1 and 4E-BP1, a process essential for DC protein synthesis. As active mTOR signaling regulates autophagy, we examined whether mf exposure alters autophagy-associated processes. mf-induced autophagy was reflected in marked upregulation of phosphorylated Beclin 1, known to play an important role in both autophagosome formation and autolysosome fusion, in induction of LC3II, a marker of autophagosome formation, and in induced degradation of p62, a ubiquitin-binding protein that aggregates protein in autophagosomes and is degraded upon autophagy that was reduced significantly by mf exposure and by rapamycin. Together, these results suggest that Brugia malayi mf employ mechanisms of metabolic modulation in DC to influence the regulation of the host immune response by downregulating mTOR signaling, resulting in increased autophagy. Whether this is a result of the parasite-secreted rapamycin homolog is currently under study.


Autophagy/physiology , Brugia malayi/parasitology , Dendritic Cells/parasitology , Microfilariae/physiology , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagosomes/metabolism , Autophagosomes/parasitology , Beclin-1/metabolism , Cell Cycle Proteins , Dendritic Cells/metabolism , Down-Regulation/physiology , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Humans , Lysosomes/metabolism , Lysosomes/parasitology , Monocytes/metabolism , Monocytes/parasitology , Phosphoproteins/metabolism , Phosphorylation/physiology , Proteomics/methods , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/physiology , Ubiquitin/metabolism , Up-Regulation/physiology
10.
Infect Immun ; 82(11): 4438-46, 2014 Nov.
Article En | MEDLINE | ID: mdl-25114121

To characterize the function and plasticity of the major human circulating monocyte populations and to explore their role in systemic helminth infection, highly purified (by flow-based sorting) human monocyte subsets (CD14(hi)/CD16(neg) [classical], CD14(+ or hi)/CD16(med) [intermediate], and CD14(neg)/CD16(hi) [nonclassical]) were examined at homeostasis and after activation. Among these three subsets the classical and intermediate subsets were found to be the major sources of inflammatory and regulatory cytokines, as well as cytokines/chemokines associated with alternative activation, whereas the nonclassical and classical populations demonstrated an ability to transmigrate through endothelial monolayers. Moreover, it was primarily the classical subset that was the most efficient in promoting autologous T cell proliferation. The distribution of these subsets changed in the context of a systemic helminth (Wuchereria bancrofti) infection such that patent infection altered the frequency and distribution of these monocyte subsets with the nonclassical monocytes being expanded (almost 2-fold) in filarial infection. To understand further the filarial/monocyte interface, in vitro modeling demonstrated that the classical subset internalized filarial antigens more efficiently than the other two subsets but that the parasite-driven regulatory cytokine interleukin-10 was exclusively coming from the intermediate subset. Our data suggest that monocyte subsets have a differential function at homeostasis and in response to helminth parasites.


Filariasis/immunology , Homeostasis/physiology , Monocytes/classification , Monocytes/physiology , Adult , Animals , Brugia malayi , Case-Control Studies , Cell Movement , Cells, Cultured , Female , Gerbillinae , Humans , Interferon-gamma/pharmacology , Interleukin-4 , Lipopolysaccharides/pharmacology , Male , Middle Aged , Young Adult
11.
Adv Exp Med Biol ; 785: 49-56, 2013.
Article En | MEDLINE | ID: mdl-23456837

Lymphatic filariasis is a mafor tropical disease affecting approximately 120 million people worldwide. Patent infection, by and large, is clinically asymptomatic but is associated with the inability of T cells to proliferate or produce IFN-γ in response to parasite antigen. Monocyte dysfunction is one hypothesis felt to explain the lack of an antigen-specific T cell response. In fact, monocytes from filaria-infected individuals have been shown to be studded with internalized filarial antigens. Understanding how the phenotype and the function of these monocytes are altered through the internalization of these parasite antigens is one of the areas our laboratory has focused on. In fact, the existence and/or function of alternatively activated macrophages in murine models of filarial infections have been extensively studied. Whether this population of macrophages can be induced in human filarial infections is the main focus of this review.


Antigens, Helminth/immunology , Brugia malayi/immunology , Elephantiasis, Filarial/immunology , Macrophages/immunology , Monocytes/immunology , T-Lymphocytes/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Elephantiasis, Filarial/parasitology , Host Specificity , Host-Parasite Interactions , Humans , Macrophage Activation , Macrophages/parasitology , Macrophages/pathology , Mice , Monocytes/parasitology , Monocytes/pathology , T-Lymphocytes/parasitology , T-Lymphocytes/pathology
12.
Infect Immun ; 81(5): 1420-9, 2013 May.
Article En | MEDLINE | ID: mdl-23429540

Filarial infection is initiated by mosquito-derived third-stage larvae (L3) deposited on the skin that transit through the epidermis, which contains Langerhans cells (LC) and keratinocytes (KC), among other cells. This earliest interaction between L3 and the LC likely conditions the priming of the immune system to the parasite. To determine the nature of this interaction, human LC (langerin(+) E-cadherin(+) CD1a(+)) were generated in vitro and exposed to live L3. LC exposed to live L3 for 48 h showed no alterations in the cell surface markers CD14, CD86, CD83, CD207, E-cadherin, CD80, CD40, and HLA-DR or in mRNA expression of inflammation-associated genes, such as those for interleukin 18 (IL-18), IL-18BP, and caspase 1. In contrast to L3, live tachyzoites of Toxoplasma gondii, an intracellular parasite, induced production of CXCL9, IP-10, and IL-6 in LC. Furthermore, preexposure of LC to L3 did not alter Toll-like receptor 3 (TLR3)- or TLR4-mediated expression of the proinflammatory cytokines IL-1ß, gamma interferon (IFN-γ), IL-6, or IL-10. Interestingly, cocultures of KC and LC produced significantly more IL-18, IL-1α, and IL-8 than did cultures of LC alone, although exposure of the cocultures to live L3 did not result in altered cytokine production. Microarray examination of ex vivo LC from skin blisters that were exposed to live L3 also showed few significant changes in gene expression compared with unexposed blisters, further underscoring the relatively muted response of LC to L3. Our data suggest that failure by LC to initiate an inflammatory response to the invasive stage of filarial parasites may be a strategy for immune evasion by the filarial parasite.


Brugia malayi/immunology , Immune Evasion/immunology , Immunity, Innate , Langerhans Cells/immunology , Animals , Antigens, CD/analysis , Antigens, CD/immunology , Cadherins/metabolism , Caspase 1/metabolism , Cytokines/metabolism , HLA-DR Antigens/analysis , Humans , Langerhans Cells/metabolism , RNA, Messenger/metabolism , Skin/immunology , Skin/parasitology
13.
Infect Immun ; 79(10): 3957-65, 2011 Oct.
Article En | MEDLINE | ID: mdl-21788379

Human monocytes from patients with patent filarial infections are studded with filarial antigen and express markers associated with alternative activation of macrophages (MΦ). To explore the role of filaria-derived parasite antigen in differentiation of human monocytes, cells were exposed to microfilariae (mf) of Brugia malayi, and their phenotypic and functional characteristics were compared with those of monocytes exposed to factors known to generate either alternatively (interleukin-4 [IL-4]) or classically (macrophage colony-stimulating factor [MCSF]) activated MΦ. IL-4 upregulated mRNA expression of CCL13, CCL15, CCL17, CCL18, CCL22, CLEC10A, MRC1, CADH1, CD274, and CD273 associated with alternative activation of MΦ but not arginase 1. IL-4-cultured monocytes had a diminished ability to promote proliferation of both CD4(+) and CD8(+) T cells compared to that of unexposed monocytes. Similar to results with IL-4, exposure of monocytes to live mf induced upregulation of CCL15, CCL17, CCL18, CCL22, CD274, and CD273 and downregulation of Toll-like receptor 3 (TLR3), TLR5, and TLR7. In contrast to results with MCSF-cultured monocytes, exposure of monocytes to mf resulted in significant inhibition of the phagocytic ability of these cells to the same degree as that seen with IL-4. Our data suggest that short exposure of human monocytes to IL-4 induces a phenotypic characteristic of alternative activation and that secreted filarial products skew monocytes similarly.


Antigens, Helminth/immunology , Brugia malayi/immunology , Cell Differentiation , Interleukin-4/immunology , Monocytes/immunology , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Down-Regulation , Filariasis/immunology , Filariasis/parasitology , Humans , Interleukin-4/metabolism , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Microfilariae/immunology , Microfilariae/metabolism , Monocytes/cytology , Phenotype , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Up-Regulation
14.
Proc Natl Acad Sci U S A ; 108(23): 9649-54, 2011 Jun 07.
Article En | MEDLINE | ID: mdl-21606368

Global proteomic analyses of pathogens have thus far been limited to unicellular organisms (e.g., protozoa and bacteria). Proteomic analyses of most eukaryotic pathogens (e.g., helminths) have been restricted to specific organs, specific stages, or secretomes. We report here a large-scale proteomic characterization of almost all the major mammalian stages of Brugia malayi, a causative agent of lymphatic filariasis, resulting in the identification of more than 62% of the products predicted from the Bm draft genome. The analysis also yielded much of the proteome of Wolbachia, the obligate endosymbiont of Bm that also expressed proteins in a stage-specific manner. Of the 11,610 predicted Bm gene products, 7,103 were definitively identified from adult male, adult female, blood-borne and uterine microfilariae, and infective L3 larvae. Among the 4,956 gene products (42.5%) inferred from the genome as "hypothetical," the present study was able to confirm 2,336 (47.1%) as bona fide proteins. Analysis of protein families and domains coupled with stage-specific expression highlight the important pathways that benefit the parasite during its development in the host. Gene set enrichment analysis identified extracellular matrix proteins and those with immunologic effects as enriched in the microfilarial and L3 stages. Parasite sex- and stage-specific protein expression identified those pathways related to parasite differentiation and demonstrates stage-specific expression by the Bm endosymbiont Wolbachia as well.


Bacterial Proteins/analysis , Brugia malayi/metabolism , Helminth Proteins/analysis , Proteome/analysis , Proteomics/methods , Wolbachia/metabolism , Animals , Bacterial Proteins/classification , Brugia malayi/growth & development , Brugia malayi/microbiology , Chromatography, Liquid/methods , Cluster Analysis , Female , Filariasis/parasitology , Helminth Proteins/classification , Host-Pathogen Interactions , Humans , Larva/growth & development , Larva/metabolism , Larva/microbiology , Life Cycle Stages , Male , Proteome/classification , Symbiosis , Tandem Mass Spectrometry , Wolbachia/physiology
15.
J Immunol ; 185(10): 6364-72, 2010 Nov 15.
Article En | MEDLINE | ID: mdl-20956349

APC dysfunction has been postulated to mediate some of the parasite-specific T cell unresponsiveness seen in patent filarial infection. We have shown that live microfilariae of Brugia malayi induce caspase-dependent apoptosis in human monocyte-derived dendritic cells (DCs) in vitro. This study addresses whether apoptosis observed in vitro extends to patent filarial infections in humans and is reflected in the number of circulating myeloid DCs (mDCs; CD11c(-)CD123(lo)) in peripheral blood of infected microfilaremic individuals. Utilizing flow cytometry to identify DC subpopulations (mDCs and plasmacytoid DCs [pDCs]) based on expression of CD11c and CD123, we found a significant increase in numbers of circulating mDCs (CD11c(+)CD123(lo)) in filaria-infected individuals compared with uninfected controls from the same filaria-endemic region of Mali. Total numbers of pDCs, monocytes, and lymphocytes did not differ between the two groups. To investigate potential causes of differences in mDC numbers between the two groups, we assessed chemokine receptor expression on mDCs. Our data indicate that filaria-infected individuals had a lower percentage of circulating CCR1(+) mDCs and a higher percentage of circulating CCR5(+) mDCs and pDCs. Finally, live microfilariae of B. malayi were able to downregulate cell-surface expression of CCR1 on monocyte-derived DCs and diminish their calcium flux in response to stimulation by a CCR1 ligand. These findings suggest that microfilaria are capable of altering mDC migration through downregulation of expression of some chemokine receptors and their signaling functions. These observations have major implications for regulation of immune responses to these long-lived parasites.


Dendritic Cells/immunology , Filariasis/immunology , Receptors, CCR1/biosynthesis , Adult , Animals , Brugia malayi/immunology , Cell Separation , Chemotaxis, Leukocyte/immunology , Clinical Trials as Topic , Dendritic Cells/metabolism , Dipetalonema Infections/immunology , Female , Filariasis/blood , Flow Cytometry , Humans , Male , Mansonella , Mansonelliasis/blood , Mansonelliasis/immunology , Middle Aged , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptors, CCR1/immunology , Reverse Transcriptase Polymerase Chain Reaction , Wuchereria bancrofti/immunology
16.
PLoS Negl Trop Dis ; 3(4): e410, 2009.
Article En | MEDLINE | ID: mdl-19352421

Relatively little is known about the filarial proteins that interact with the human host. Although the filarial genome has recently been completed, protein profiles have been limited to only a few recombinants or purified proteins of interest. Here, we describe a large-scale proteomic analysis using microcapillary reverse-phase liquid chromatography-tandem-mass spectrometry to identify the excretory-secretory (ES) products of the L3, L3 to L4 molting ES, adult male, adult female, and microfilarial stages of the filarial parasite Brugia malayi. The analysis of the ES products from adult male, adult female, microfilariae (Mf), L3, and molting L3 larvae identified 852 proteins. Annotation suggests that the functional and component distribution was very similar across each of the stages studied; however, the Mf contributed a higher proportion to the total number of identified proteins than the other stages. Of the 852 proteins identified in the ES, only 229 had previous confirmatory expressed sequence tags (ESTs) in the available databases. Moreover, this analysis was able to confirm the presence of 274 "hypothetical" proteins inferred from gene prediction algorithms applied to the B. malayi (Bm) genome. Not surprisingly, the majority (160/274) of these "hypothetical" proteins were predicted to be secreted by Signal IP and/or SecretomeP 2.0 analysis. Of major interest is the abundance of previously characterized immunomodulatory proteins such as ES-62 (leucyl aminopeptidase), MIF-1, SERPIN, glutathione peroxidase, and galectin in the ES of microfilariae (and Mf-containing adult females) compared to the adult males. In addition, searching the ES protein spectra against the Wolbachia database resulted in the identification of 90 Wolbachia-specific proteins, most of which were metabolic enzymes that have not been shown to be immunogenic. This proteomic analysis extends our knowledge of the ES and provides insight into the host-parasite interaction.


Brugia malayi/chemistry , Brugia malayi/growth & development , Helminth Proteins/metabolism , Host-Pathogen Interactions , Proteome/analysis , Animals , Chromatography, Liquid , Erythrocytes , Female , Male , Mass Spectrometry
17.
Cancer Immunol Immunother ; 58(11): 1809-18, 2009 Nov.
Article En | MEDLINE | ID: mdl-19319531

This study demonstrates that CD8+ T cells in the tumor microenvironment display reduced functionality and hyporesponsiveness. TGF-beta contributed markedly to the tumor-infiltrating CD8+ T cells' (TILs) reduced functionality, which could be reversed using a small molecule TGF-beta inhibitor. Upon T-cell receptor (TCR) activation, the activation of ITK and ERK kinases were reduced in CD8+ TILs, as compared to splenic CD8+ T cells: TGF-beta inhibitor could reverse this phenomenon. This study demonstrates for the first time the association of the Spred-1 gene, an inhibitor of the Ras/MAPK pathway, with CD8+ TILs and TGF-beta activity. Spred-1 was upregulated in CD8+ TILs and TGF-beta enhanced the expression of Spred-1 in effector/memory CD8+ T cells and not in rested/memory CD8+ T cells. Based on these findings, this study supports the hypothesis that TGF-beta mediates an inhibitory mechanism on CD8+ TILs involving TCR-signaling blockade and the upregulation of Spred-1, thus implicating Spred-1 as a potential new target for future anti-tumor immune studies.


CD8-Positive T-Lymphocytes/physiology , Lymphocytes, Tumor-Infiltrating/physiology , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/physiology , Repressor Proteins/physiology , Transforming Growth Factor beta/physiology , Adaptor Proteins, Signal Transducing , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Immune Tolerance , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Phosphorylation , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors
18.
Immunol Res ; 43(1-3): 252-63, 2009.
Article En | MEDLINE | ID: mdl-18982454

Helminth (worm) infections are major public health problems that have important socioeconomic consequences for the more than 2 billion infected individuals. Chronicity (their hallmark) can lead to anemia (in hookworm infection), river blindness (onchcerciasis), cirrhosis (schistosomiasis), and elephantiasis (lymphatic filariasis). Although there have been many studies examining innate immune responses (including TLR expression and function) in response to intracellular pathogens, fewer have examined the interaction of the multicellular helminth parasites and the innate immune system. This review will focus on two "systemic" helminth parasitic infections (lymphatic filariasis and schistosomiasis) and the regulation of TLRs that may contribute to infection outcome.


Brugia malayi/immunology , Elephantiasis, Filarial/immunology , Schistosoma/immunology , Schistosomiasis/immunology , Toll-Like Receptors/immunology , Wuchereria bancrofti/immunology , Animals , Cytokines/immunology , Humans , Toll-Like Receptors/agonists
19.
J Immunol ; 181(10): 7081-9, 2008 Nov 15.
Article En | MEDLINE | ID: mdl-18981128

Dysregulation of professional APC has been postulated as a major mechanism underlying Ag-specific T cell hyporesponsiveness in patients with patent filarial infection. To address the nature of this dysregulation, dendritic cells (DC) and macrophages generated from elutriated monocytes were exposed to live microfilariae (mf), the parasite stage that circulates in blood and is responsible for most immune dysregulation in filarial infections. DC exposed to mf for 24-96 h showed a marked increase in cell death and caspase-positive cells compared with unexposed DC, whereas mf exposure did not induce apoptosis in macrophages. Interestingly, 48-h exposure of DC to mf induced mRNA expression of the proapoptotic gene TRAIL and both mRNA and protein expression of TNF-alpha. mAb to TRAIL-R2, TNF-R1, or TNF-alpha partially reversed mf-induced cell death in DC, as did knocking down the receptor for TRAIL-R2 using small interfering RNA. The mf also induced gene expression of BH3-interacting domain death agonist and protein expression of cytochrome c in DC; mf-induced cleavage of BH3-interacting domain death agonist could be shown to induce release of cytochrome c, leading to activation of caspase 9. Our data suggest that mf induce DC apoptosis in a TRAIL- and TNF-alpha-dependent fashion.


Apoptosis/immunology , Dendritic Cells/immunology , Filariasis/immunology , Microfilariae/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , BH3 Interacting Domain Death Agonist Protein/biosynthesis , Brugia malayi/immunology , Cytochromes c/biosynthesis , Dendritic Cells/metabolism , Flow Cytometry , Gene Expression , Gene Expression Regulation , Humans , Immunoblotting , Macrophages/immunology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
20.
Blood ; 112(4): 1290-8, 2008 Aug 15.
Article En | MEDLINE | ID: mdl-18541719

Patent lymphatic filariasis is characterized by antigen-specific T-cell unresponsiveness with diminished IFN-gamma and IL-2 production and defects in dendritic cell (DC) function. Because Toll-like receptors (TLRs) play an important role in pathogen recognition and TLR expression is diminished on B and T cells of filaria-infected individuals, we examined the effect of live microfilariae (mf) on expression and function of TLRs in human DCs. We show that mf-exposed monocyte-derived human DCs (mhDCs) demonstrate marked diminution of TLR3 and TLR4 mRNA expression compared with mf-unexposed mhDCs that translated into loss of function in response to appropriate TLR ligands. Exposure to mf significantly down-regulated production of IFN-alpha, MIP-1alpha, IL-12p70, and IL-1alpha following activation with poly I:C, and of IL-12p40 following activation with poly I:C or LPS. mRNA expression of MyD88, the adaptor molecule involved in TLR4 signaling, was significantly diminished in mhDCs after exposure to mf. Moreover, mf interfered with NF-kappaB activation (particularly p65 and p50) following stimulation with poly I:C or LPS. These data suggest that mf interfere with mhDC function by altering TLR expression and interfering with both MyD88-dependent signaling and a pathway that ultimately diminishes NF-kappaB activity. This down-regulated NF-kappaB activity impairs mhDC-produced cytokines needed for full T-cell activation.


Dendritic Cells/parasitology , Helminths/immunology , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/immunology , Down-Regulation , Humans , Lymphocyte Activation , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , RNA, Messenger/analysis , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics
...