Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
NPJ Vaccines ; 6(1): 86, 2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34145272

ABSTRACT

Activation of antigen-specific T-lymphocyte responses may be needed to cure disorders caused by chronic infection with low-risk human papillomavirus (lrHPV). Safe and effective adjuvant therapies for such disorders are needed. The safety and efficacy of a novel gorilla adenovirus vaccine expressing a protein designed to elicit immune responses directed against HPV6 and HPV11, PRGN-2012, was studied using in vitro stimulation of T lymphocytes from patients with recurrent respiratory papillomatosis, in vivo vaccination studies, and therapeutic studies in mice bearing tumors expressing lrHPV antigen. PRGN-2012 treatment induces lrHPV antigen-specific responses in patient T lymphocytes. Vaccination of wild-type mice induces E6-specific T-lymphocyte responses without toxicity. In vivo therapeutic vaccination of mice bearing established HPV6 E6 expressing tumors results in HPV6 E6-specific CD8+ T-lymphocyte immunity of sufficient magnitude to induce tumor growth delay. The clinical study of PRGN-2012 in patients with disorders caused by chronic infection with lrHPV is warranted.

2.
Retrovirology ; 17(1): 17, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32615983

ABSTRACT

BACKGROUND: Heterosexual transmission remains the main route of HIV-1 transmission and female genital tract (FGT) inflammation increases the risk of infection. However, the mechanism(s) by which inflammation facilitates infection is not fully understood. In rhesus macaques challenged with simian immunodeficiency virus, dendritic cell (DC) mediated recruitment of CD4+ T cells to the FGT was critical for infection. The aim of this study was to delineate the mechanisms underlying DC-mediated HIV infection by comparing chemokine and pro-inflammatory cytokine production in response to transmitted founder (TF) and chronic infection (CI) Envelope (Env) pseudotyped viruses (PSV). RESULTS: Monocyte-derived DCs (MDDCs) were stimulated with PSV and recombinant gp140 representing matched TF and CI pairs of four individuals and cytokine secretion measured by multiplex immuno-assay. We found that 4/9 Env induced robust MDDC inflammatory responses and of those, three were cloned from TFs. Overall, TF Env induced MDDCs from healthy donors to secrete higher concentrations of inflammatory cytokines and chemokines than those from CI, suggesting TF Env were better inducers of inflammation. Assessing the signalling pathway associated with inflammatory cytokines, we found that PSV of matched TF and CI variants and a gp140 clone activated ERK and JNK to similar levels. Recombinant soluble DC-SIGN inhibited cytokine release and activation of ERK by PSV, suggesting that Env-DC-SIGN binding was partly involved in MDDC stimulation. Therefore, Env clones might differentially stimulate MDDC immune responses via alternative, yet unidentified signalling pathways. CONCLUSION: Overall, this could suggest that the genetics of the virus itself influences inflammatory responses during HIV infection. In the absence of pre-existing infections, induction of greater inflammatory response by TFs might favour virus survival within the healthy FGT by driving an influx of target cells to sites of infection while suppressing immune responses via IL-10.


Subject(s)
Dendritic Cells/immunology , HIV Infections/virology , HIV-1/physiology , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Cytokines/metabolism , Female , Humans , Lectins, C-Type/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Monocytes/immunology , Receptors, Cell Surface/metabolism , Signal Transduction , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/metabolism
3.
PLoS Negl Trop Dis ; 12(3): e0006345, 2018 03.
Article in English | MEDLINE | ID: mdl-29579050

ABSTRACT

The disease burden of Wuchereria bancrofti and Plasmodium falciparum malaria is high, particularly in Africa, and co-infection is common. However, the effects of filarial infection on the risk of severe malaria are unknown. We used the remaining serum samples from a large cohort study in Muheza, Tanzania to describe vector-borne filarial sero-reactivity among young children and to identify associations between exposure to filarial parasites and subsequent severe malaria infections. We identified positive filarial antibody responses (as well as positive antibody responses to Strongyloides stercoralis) among infants as young as six months. In addition, we found a significant association between filarial seropositivity at six months of age and subsequent severe malaria. Specifically, infants who developed severe malaria by one year of age were 3.9 times more likely (OR = 3.9, 95% CI: 1.2, 13.0) to have been seropositive for filarial antigen at six months of age compared with infants who did not develop severe malaria.


Subject(s)
Antibodies, Helminth/blood , Brugia malayi/immunology , Filariasis/epidemiology , Malaria/epidemiology , Wuchereria bancrofti/immunology , Animals , Antigens, Helminth/immunology , Child , Child, Preschool , Cohort Studies , Coinfection/epidemiology , Cytokines/blood , Female , Humans , Infant , Malaria/blood , Malaria/immunology , Malaria/parasitology , Male , Prevalence , Risk Factors , Seroepidemiologic Studies , Severity of Illness Index , Strongyloides stercoralis/immunology , Tanzania/epidemiology , Wuchereria bancrofti/isolation & purification
4.
Clin Infect Dis ; 64(8): 1017-1025, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28329346

ABSTRACT

Background: Severe adverse reactions have been observed in individuals with Loa loa infection treated with either diethylcarbamazine (DEC), the drug of choice for loiasis, or ivermectin (IVM), which is used in mass drug administration programs for control of onchocerciasis and lymphatic filariasis in Africa. In this study, posttreatment clinical and immunologic reactions were compared following single-dose therapy with DEC or IVM to assess whether these reactions have the same underlying pathophysiology. Methods: Twelve patients with loiasis and microfilarial counts <2000 mf/mL were randomized to receive single-dose DEC (8 mg/kg) or IVM (200 µg/kg). Clinical and laboratory assessments were performed at 4, 8, 24, 48, and 72 hours and 5, 7, 9, and 14 days posttreatment. Results: Posttreatment adverse events were similar following DEC or IVM, but peaked earlier in subjects who received DEC, consistent with a trend toward more rapid and complete microfilarial clearance in the DEC group. After a transient rise (post-IVM) or fall (post-DEC) in the first 24 hours posttreatment, the eosinophil count rose significantly in both groups, peaking at day 5 in the DEC group and day 9 in the IVM group. Serum interleukin 5 levels and eosinophil activation, as assessed by surface expression of CD69 and serum levels of eosinophil granule proteins, were increased posttreatment in both groups. Conclusions: Despite differences in eosinophil and lymphocyte counts during the first 24 hours posttreatment, the overall pattern of hematologic and immunologic changes suggest that posttreatment reactions following DEC and IVM share a common pathophysiology. Clinical Trials Registration: NCT01593722.


Subject(s)
Diethylcarbamazine/adverse effects , Drug-Related Side Effects and Adverse Reactions/pathology , Filaricides/adverse effects , Ivermectin/adverse effects , Loiasis/drug therapy , Adult , Aged , Diethylcarbamazine/administration & dosage , Female , Filaricides/administration & dosage , Humans , Ivermectin/administration & dosage , Male , Middle Aged , Pilot Projects , Young Adult
5.
J Circ Biomark ; 5: 5, 2016.
Article in English | MEDLINE | ID: mdl-28936253

ABSTRACT

Recent advances in human immunology have led to the identification of novel immune cell subsets and the biological function of many of these subsets has now been identified. The recent US Food and Drug Administration approval of several immunotherapeutics for the treatment of a variety of cancer types and the results of ongoing immunotherapy clinical studies requires a more thorough interrogation of the immune system. We report here the use of flow cytometry-based analyses to identify 123 immune cell subsets of peripheral blood mononuclear cells. The use of these panels defines multiple differences in younger (< 40 years) vs. older (≥ 40 years) individuals and between aged-matched apparently healthy individuals and metastatic cancer patients, aspects not seen in the analysis of the following standard immune cell types: CD8, CD4, natural killer, natural killer-T, regulatory T, myeloid derived suppressor cells, conventional dendritic cells (DCs), plasmacytoid DCs and B cells. The use of these panels identifying 123 immune cell subsets may aid in the identification of patients who may benefit from immunotherapy, either prior to therapy or early in the immunotherapeutic regimen, for the treatment of cancer or other chronic or infectious diseases.

6.
Clin Infect Dis ; 60(1): 55-63, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25234520

ABSTRACT

BACKGROUND: Loa loa has emerged as an important public health problem due to the occurrence of immune-mediated severe posttreatment reactions following ivermectin distribution. Also thought to be immune-mediated are the dramatic differences seen in clinical presentation between infected temporary residents (TR) and individuals native to endemic regions (END). METHODS: All patients diagnosed with loiasis at the National Institutes of Health between 1976 and 2012 were included. Patients enrolled in the study underwent a baseline clinical and laboratory evaluation and had serum collected and stored. Stored pretreatment serum was used to measure filaria-specific antibody responses, eosinophil-related cytokines, and eosinophil granule proteins. RESULTS: Loa loa infection in TR was characterized by the presence of Calabar swelling (in 82% of subjects), markedly elevated eosinophil counts, and increased filaria-specific immunoglobulin G (IgG) levels; these findings were thought to reflect an unmodulated immune response. In contrast, END showed strong evidence for immune tolerance to the parasite, with high levels of circulating microfilariae, few clinical symptoms, and diminished filaria-specific IgG. The striking elevation in eosinophil counts among the TR group was accompanied by increased eosinophil granule protein levels (associated with eosinophil activation and degranulation) as well as elevated levels of eosinophil-associated cytokines. CONCLUSIONS: These data support the hypothesis that differing eosinophil-associated responses to the parasite may be responsible for the marked differences in clinical presentations between TR and END populations with loiasis.


Subject(s)
Endemic Diseases , Eosinophils/immunology , Loiasis/epidemiology , Loiasis/pathology , Adult , Animals , Antibodies, Helminth/blood , Cytokines/blood , Eosinophil Granule Proteins/analysis , Female , Humans , Loa/immunology , Loiasis/immunology , Male
7.
Eur J Immunol ; 44(7): 2036-47, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24737144

ABSTRACT

The mechanisms underlying the increase in the numbers of regulatory T (Treg) cells in chronic infection settings remain unclear. Here we have delineated the phenotype and transcriptional profiles of Treg cells from 18 filarial-infected (Fil(+) ) and 19 filarial-uninfected (Fil(-) ) subjects. We found that the frequencies of Foxp3(+) Treg cells expressing CTLA-4, GITR, LAG-3, and IL-10 were significantly higher in Fil(+) subjects compared with that in Fil(-) subjects. Foxp3-expressing Treg-cell populations in Fil(+) subjects were also more heterogeneous and had higher expression of IL-10, CCL-4, IL-29, CTLA-4, and TGF-ß than Fil(-) subjects, each of these cytokines having been implicated in immune suppression. Moreover, Foxp3-expressing Treg cells from Fil(+) subjects had markedly upregulated expression of activation-induced apoptotic genes with concomitant downregulation of those involved in cell survival. To determine whether the expression of apoptotic genes was due to Treg-cell activation, we found that the expression of CTLA-4, CDk8, RAD50, TNFRSF1A, FOXO3, and RHOA were significantly upregulated in stimulated cells compared with unstimulated cells. Taken together, our results suggest that in patent filarial infection, the expanded Treg-cell populations are heterogeneous, short-lived, activated, and express higher levels of molecules known to modulate immune responsiveness, suggesting that filarial infection is associated with high Treg-cell turnover.


Subject(s)
Filariasis/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Antigens, CD/analysis , Apoptosis , CTLA-4 Antigen/analysis , Chronic Disease , Female , Forkhead Transcription Factors/analysis , Humans , Interleukin-10/analysis , Male , Lymphocyte Activation Gene 3 Protein
8.
Front Immunol ; 4: 305, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24137161

ABSTRACT

Filarial infections in humans are chronic infections that cause significant morbidity. The chronic nature of these infections with continuous antigen release is associated with a parasite-specific T cell hypo-responsiveness that may over time also affect the immune responses to bystander antigens. Previous studies have shown the filarial parasite antigen-specific T cells hypo-responsiveness is mediated by regulatory cytokines - IL-10 and TGF-ß in particular. Recent studies have suggested that the modulated/regulated T cell responses associated with patent filarial infection may reflect an expansion of regulatory T cells (Tregs) that include both Tregs induced in peripheral circulation or pTregs and the thymus-derived Tregs or tTregs. Although much is known about the phenotype of these regulatory populations, the mechanisms underlying their expansion and their mode of action in filarial and other infections remain unclear. Nevertheless there are data to suggest that while many of these regulatory cells are activated in an antigen-specific manner the ensuing effectors of this activation are relatively non-specific and may affect a broad range of immune cells. This review will focus on the subsets and function of regulatory T cells in filarial infection.

9.
PLoS Negl Trop Dis ; 6(11): e1890, 2012.
Article in English | MEDLINE | ID: mdl-23133692

ABSTRACT

BACKGROUND: Wuchereria bancrofti (Wb) and Mansonella perstans (Mp) are blood-borne filarial parasites that are endemic in many countries of Africa, including Mali. The geographic distribution of Wb and Mp overlaps considerably with that of malaria, and coinfection is common. Although chronic filarial infection has been shown to alter immune responses to malaria parasites, its effect on clinical and immunologic responses in acute malaria is unknown. METHODOLOGY/PRINCIPAL FINDINGS: To address this question, 31 filaria-positive (FIL+) and 31 filaria-negative (FIL-) children and young adults, matched for age, gender and hemoglobin type, were followed prospectively through a malaria transmission season. Filarial infection was defined by the presence of Wb or Mp microfilariae on calibrated thick smears performed between 10 pm and 2 am and/or by the presence of circulating filarial antigen in serum. Clinical malaria was defined as axillary temperature ≥37.5°C or another symptom or sign compatible with malaria infection plus the presence of asexual malaria parasites on a thick blood smear. Although the incidence of clinical malaria, time to first episode, clinical signs and symptoms, and malaria parasitemia were comparable between the two groups, geometric mean hemoglobin levels were significantly decreased in FIL- subjects at the height of the transmission season compared to FIL+ subjects (11.4 g/dL vs. 12.5 g/dL, p<0.01). Plasma levels of IL-1ra, IP-10 and IL-8 were significantly decreased in FIL+ subjects at the time of presentation with clinical malaria (99, 2145 and 49 pg/ml, respectively as compared to 474, 5522 and 247 pg/ml in FIL- subjects). CONCLUSIONS/SIGNIFICANCE: These data suggest that pre-existent filarial infection attenuates immune responses associated with severe malaria and protects against anemia, but has little effect on susceptibility to or severity of acute malaria infection. The apparent protective effect of filarial infection against anemia is intriguing and warrants further study in a larger cohort.


Subject(s)
Anemia/pathology , Cytokines/blood , Filariasis/complications , Filariasis/pathology , Malaria/complications , Malaria/pathology , Adolescent , Anemia/etiology , Animals , Child , Child, Preschool , Female , Filariasis/immunology , Hemoglobins/analysis , Humans , Incidence , Malaria/immunology , Male , Mali , Mansonella/isolation & purification , Plasmodium/isolation & purification , Prospective Studies , Wuchereria bancrofti/isolation & purification , Young Adult
10.
Curr Opin HIV AIDS ; 7(3): 231-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22418448

ABSTRACT

PURPOSE OF REVIEW: To examine the consequences of the immune modulation seen in chronic filarial infection on responses to intracellular pathogens (and their antigens) that are often co-endemic with filarial infections, namely Plasmodium and Mycobacterium tuberculosis. RECENT FINDINGS: Much of the recent data on filaria/mycobacteria or filaria/Plasmodium co-infection has focused on the modulation of mycobacteria-specific or malaria-specific responses by chronic filarial infection. As such, filarial infections very clearly alter the magnitude and quality of the mycobacteria-specific or malaria-specific cytokine responses, responses that have been typically associated with control of these intracellular pathogens. SUMMARY: Although phylogenetically distinct, mycobacteria and Plasmodium spp. often share the same geographical niche with filarial infections. The complex interplay between filarial parasites that are associated with immunomodulation and those microbial pathogens that require a proinflammatory or unmodulated response for their control is easily demonstrable ex vivo, but whether this interplay affects disease outcome in tuberculosis or malaria remains an open question.


Subject(s)
Coinfection/immunology , Filariasis/complications , Filarioidea/immunology , Malaria, Falciparum/immunology , Tuberculosis/immunology , Animals , Coinfection/microbiology , Coinfection/parasitology , Cytokines , Filariasis/parasitology , Humans , Malaria, Falciparum/complications , Mycobacterium tuberculosis/immunology , Plasmodium falciparum/immunology , Tuberculosis/complications
11.
Eur J Immunol ; 42(3): 641-50, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22213332

ABSTRACT

In areas where polyparasitism is highly prevalent, the impact of multiple parasites on the host response is underestimated. In particular, the presence of helminth infection coincident with malaria profoundly alters the production of malaria-specific IFN-γ, IL-12p70, CXCL9, CXCL10 and CXCL11, cytokines/chemokines known to be critical in mediating malaria-specific immunity. In order to elucidate the mechanisms underlying the suppression of malaria-specific cytokines/chemokines, we assessed the expression of malaria-specific IL-12Rß1, IL-12Rß2 and interferon regulatory factor (IRF)-1 in blood obtained from 18 filaria-infected (Fil(+)) and 17 filaria-uninfected (Fil(-)) individuals in a filaria-malaria co-endemic region of Mali. We found that Fil(+) individuals had significantly lower RNA expression of IRF-1 but not IL-12Rß1 or IL-12Rß2 in response to malaria antigen stimulation. We also measured the frequency of IL-12-producing DCs from these subjects and found that Fil(+) subjects had lower frequencies of IL-12(+) mDCs after malaria antigen stimulation than did the Fil(-) subjects. Modeling these data in vitro, we found that mDCs pre-exposed to live microfilariae not only produced significantly lower levels of CXCL-9, CXCL-10, IL-12p35, IL-12p40, IL-12p19 and CXCL-11 following stimulation with malaria antigen but also markedly downregulated the expression of IRF-1, IRF-2 and IRF-3 compared with microfilaria-unexposed mDCs. siRNA-inhibition of irf-1 in mDCs downregulated the production of IL-12p70 through repression of IL-12p35. Our data demonstrate that the modulation of IRFs seen in filarial (and presumably other tissue-invasive helminths) infection underlies the suppression of malaria-specific cytokines/chemokines that play a crucial role in immunity to malaria.


Subject(s)
Brugia malayi/immunology , Filariasis/immunology , Interferon Regulatory Factor-1/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Animals , Antigens, Protozoan/immunology , Chemokine CXCL10/immunology , Chemokine CXCL11/immunology , Chemokine CXCL9/immunology , Dendritic Cells/immunology , Down-Regulation , Filariasis/parasitology , Flow Cytometry , Humans , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-1/genetics , Malaria, Falciparum/parasitology , Mali , RNA/chemistry , RNA/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction
12.
J Immunol ; 186(8): 4725-33, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21411732

ABSTRACT

The mechanisms underlying the modulation of both the malaria-specific immune response and the course of clinical malaria in the context of concomitant helminth infection are poorly understood. We used multiparameter flow cytometry to characterize the quality and the magnitude of malaria-specific T cell responses in filaria-infected and -uninfected individuals with concomitant asymptomatic Plasmodium falciparum malaria in Mali. In comparison with filarial-uninfected subjects, filarial infection was associated with higher ex vivo frequencies of CD4(+) cells producing IL-4, IL-10, and IL-17A (p = 0.01, p = 0.001, and p = 0.03, respectively). In response to malaria Ag stimulation, however, filarial infection was associated with lower frequencies of CD4(+) T cells producing IFN-γ, TNF-α, and IL-17A (p < 0.001, p = 0.04, and p = 0.04, respectively) and with higher frequencies of CD4(+)IL10(+)T cells (p = 0.0005). Importantly, filarial infection was associated with markedly lower frequencies of malaria Ag-specific Th1 (p < 0.0001), Th17 (p = 0.012), and "TNF-α" (p = 0.0008) cells, and a complete absence of malaria-specific multifunctional Th1 cells. Filarial infection was also associated with a marked increase in the frequency of malaria-specific adaptive regulatory T/Tr1 cells (p = 0.024), and the addition of neutralizing anti-IL-10 Ab augmented the amount of Th1-associated cytokine produced per cell. Thus, among malaria-infected individuals, concomitant filarial infection diminishes dramatically the frequencies of malaria-specific Th1 and Th17 T cells, and alters the quality and magnitude of malaria-specific T cell responses.


Subject(s)
Filariasis/immunology , Malaria, Falciparum/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adolescent , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Child , Comorbidity , Female , Filariasis/epidemiology , Filariasis/parasitology , Filarioidea/immunology , Flow Cytometry , Humans , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Male , Mali/epidemiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
13.
J Immunol ; 185(10): 6364-72, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20956349

ABSTRACT

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.


Subject(s)
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
14.
J Immunol ; 184(9): 5375-82, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20357251

ABSTRACT

Despite the well-documented immune suppression associated with human helminth infections, studies characterizing the immune response at the single-cell level are scanty. We used multiparameter flow cytometry to characterize the type of effector (Th1, Th2, and Th17) and regulatory (natural T regulatory cells [nTregs] and adaptive Treg cells [aTreg/type 1 regulatory cells (Tr1s)]) CD4(+) and CD8(+) T cells in filaria-infected (Fil(+)) and -uninfected (Fil(-)) individuals at homeostasis (in the absence of stimulation). Frequencies of CD4(+) lymphocytes spontaneously producing IL-4, IL-10, and IL-17A were significantly higher in Fil(+), as were those of IL-10(+)/IL-4(+) double-producing CD4(+) cells. Interestingly, frequencies of Th17 and aTreg/Tr1s but not classical Th1 or Th2 cells were significantly increased in Fil(+) compared to Fil(-) individuals. Although the frequency of nTreg was increased in Fil(+), IL-10 was overwhelmingly produced by CD4(+)CD25(-) cells. Moreover, the concentration of IL-10 produced spontaneously in vitro strongly correlated with the integrated geometric mean fluorescence intensity of IL-10-producing aTreg/Tr1s in Fil(+). Together, these data show that at steady state, IL-10-producing aTreg/Tr1 as well as nTreg and effector Th17 CD4(+) cells are expanded in vivo in human filarial infections. Moreover, we have established baseline ex vivo frequencies of effector and Tregs at homeostasis at a population level.


Subject(s)
Adaptive Immunity , Elephantiasis, Filarial/immunology , Homeostasis/immunology , Mansonelliasis/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Cytokines/biosynthesis , Enterobiasis/immunology , Enterobius/immunology , Humans , Hymenolepiasis/immunology , Hymenolepis nana/immunology , Mansonella/immunology , Microfilariae/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/parasitology , Th2 Cells/metabolism , Th2 Cells/parasitology , Wuchereria bancrofti/immunology
15.
J Immunol ; 183(2): 916-24, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19561105

ABSTRACT

The effect of filarial infections on malaria-specific immune responses was investigated in Malian villages coendemic for filariasis (Fil) and malaria. Cytokines were measured from plasma and Ag-stimulated whole blood from individuals with Wuchereria bancrofti and/or Mansonella perstans infections (Fil(+); n = 19) and those without evidence of filarial infection (Fil(-); n = 19). Plasma levels of IL-10 (geometric mean [GM], 22.8 vs 10.4) were higher in Fil(+) compared with Fil(-), whereas levels of IFN-inducible protein (IP)-10 were lower in Fil(+) (GM, 66.3 vs 110.0). Fil(+) had higher levels of spontaneously secreted IL-10 (GM, 59.3 vs 6.8 pg/ml) and lower levels of IL-2 (1.0 vs 1.2 pg/ml) than did Fil(-). Although there were no differences in levels of Staphylococcus aureus enterotoxin B-induced cytokines between the two groups, Fil(+) mounted lower IL-12p70 (GM, 1.11 vs 3.83 pg/ml; p = 0.007), IFN-gamma (GM, 5.44 vs 23.41 pg/ml; p = 0.009), and IP-10 (GM, 29.43 vs 281.7 pg/ml; p = 0.007) responses following malaria Ag (MalAg) stimulation compared with Fil(-). In contrast, Fil(+) individuals had a higher MalAg-specific IL-10 response (GM, 7318 pg/ml vs 3029 pg/ml; p = 0.006) compared with those without filarial infection. Neutralizing Ab to IL-10 (but not to TGFbeta) reversed the down-regulated MalAg-specific IFN-gamma and IP-10 (p < 0.001) responses in Fil(+). Together, these data demonstrate that filarial infections modulate the Plasmodium falciparum-specific IL-12p70/IFN-gamma secretion pathways known to play a key role in resistance to malaria and that they do so in an IL-10-dependent manner.


Subject(s)
Cytokines/immunology , Filariasis/complications , Interleukin-10/blood , Malaria, Falciparum/complications , Animals , Cross Reactions/immunology , Cytokines/metabolism , Endemic Diseases , Filariasis/immunology , Humans , Immunity , Interferon-gamma/metabolism , Interleukin-12/metabolism , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology
16.
J Immunol ; 178(5): 2770-7, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17312120

ABSTRACT

Plasmodium falciparum infection during pregnancy can lead to the transplacental passage of malarial Ags that are capable of inducing acquired immune responses in the fetus. Studies have identified cytokines produced by malaria-specific cord blood (CB) T cells, but information on fetal B cells is limited. Thus, CB mononuclear cells from 120 Cameroonian newborns were cultured for 7 days in vitro and supernatants were assessed by ELISA for Abs to an extract of malarial schizonts (MA), recombinant apical merozoite Ag 1 (AMA-1), the 42-kDa C-terminal region of merozoite surface protein 1 (MSP-1(42)), a B epitope of ring-infected erythrocyte surface Ag (RESA), and the dominant B epitope of the circumsporozoite protein (CSP). Only 12% of supernatants contained IgM to MA but 78% had IgG to one or more malarial Ags, with 53% having IgG to AMA-1, 38% to MSP-1(42), 3% to RESA, and 0% to CSP. The Abs to AMA-1 and MSP-1(42) were predominantly IgG1 and IgG3. CB mononuclear cells were also tested for the ability to secrete cytokines in response to MA and a pool of conserved MSP-1 T cell epitopes. Among the Ag-reactive samples, 39.3% produced only Th2-type cytokines, whereas 60.6% produced a combination of Th1- and Th2-type cytokines. Although a Th2 bias was observed, the in utero cytokine environment was adequate to support isotype switching to cytophilic IgGs, the isotypes that are protective in adults. Because many infants living in a low transmission area are born with malaria-specific B and T cells, the influence of in utero priming on neonatal immunity merits further investigation.


Subject(s)
Antigens, Protozoan/immunology , Fetus/immunology , Malaria, Falciparum/immunology , Maternal-Fetal Exchange/immunology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Animals , Antibodies, Protozoan/immunology , Cameroon , Cytokines/immunology , Endemic Diseases , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Female , Fetal Blood/immunology , Humans , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Male , Pregnancy , Th1 Cells/immunology , Th2 Cells/immunology
17.
Infect Immun ; 72(9): 5267-73, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15322022

ABSTRACT

Sequestration of Plasmodium falciparum parasites within the placenta often leads to an accumulation of macrophages within the intervillous space and increased production of tumor necrosis factor alpha (TNF-alpha), a cytokine associated with placental pathology and poor pregnancy outcomes. P. falciparum glycosylphosphatidylinositol (GPI) anchors have been shown to be the major parasite component that induces TNF-alpha production by monocytes and macrophages. Antibodies against P. falciparum GPI (anti-PfGPI), however, can inhibit the induction of TNF-alpha and inflammation. Thus, the study was undertaken to determine whether anti-PfGPI antibodies down-regulate inflammatory-type changes in the placentas of women with malaria. Anti-PfGPI immunoglobulin M (IgM) and IgG levels were measured in 380 pregnant women with or without placental malaria, including those who delivered prematurely and at term. Results showed that anti-PfGPI antibody levels increased with gravidity and age and that malaria infection boosted anti-PfGPI antibodies in pregnant women. However, no association was found between anti-PfGPI antibodies and placental TNF-alpha levels or the presence of acute or chronic placental malaria. Furthermore, anti-PfGPI antibody levels were similar in women with preterm and full-term deliveries and were not associated with an increase in infant birth weight. Thus, these results fail to support a strong role for anti-PfGPI antibodies in the prevention of chronic placental malaria infections and malaria-associated poor birth outcomes.


Subject(s)
Antibodies, Protozoan/blood , Glycosylphosphatidylinositols/immunology , Placenta/parasitology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Pregnancy Outcome , Adult , Age Factors , Animals , Cameroon , Delivery, Obstetric , Female , Gravidity , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Infant, Newborn , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Placenta/immunology , Placenta Diseases/parasitology , Plasmodium falciparum/chemistry , Pregnancy , Tumor Necrosis Factor-alpha/metabolism
18.
J Infect Dis ; 188(7): 1074-82, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14513430

ABSTRACT

Plasmodium falciparum-infected erythrocytes often are sequestered in the placenta and stimulate the accumulation of maternal mononuclear cells. In this study, the role that chemokines and cytokines play in mediating the inflammatory response was investigated. Placental parasites elicited a statistically significant increase in the levels of interferon (IFN)-gamma, tumor necrosis factor (TNF)-alpha, and interleukin (IL)-10, in plasma collected from the intervillous space. Explants of fetal tissue from malaria-positive placentas also secreted significantly enhanced amounts of IFN-gamma. Culture supernatant of maternal intervillous leukocytes obtained from infected placentas contained significantly higher levels of TNF-alpha, IL-10, monocyte chemotactic protein-1, macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and IFN-gamma inducible protein-10 than did cultures of white blood cells obtained from uninfected placentas. Taken together, these results show that both fetal and maternal cells secrete inflammatory and immunoregulatory cytokines in response to P. falciparum and suggest that beta-chemokines produced by maternal cells contribute to the accumulation of macrophages in the intervillous space.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Malaria, Falciparum/immunology , Placenta/immunology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Adult , Animals , Cameroon , Enzyme-Linked Immunosorbent Assay , Female , Fetus/parasitology , Hematocrit , Humans , Immunohistochemistry , Leukocyte Count , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Parasitemia/blood , Parasitemia/immunology , Parasitemia/parasitology , Placenta/parasitology , Plasmodium falciparum/metabolism , Pregnancy , Pregnancy Complications, Parasitic/blood , Pregnancy Complications, Parasitic/parasitology
19.
Am J Trop Med Hyg ; 69(6): 574-81, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14740871

ABSTRACT

The prevalence of pre-term deliveries (PTDs) is increased in women who become infected with Plasmodium falciparum during pregnancy. Because prematurity is a risk factor for newborns, it is important to identify conditions that contribute to malaria-associated PTDs. Plasmodium falciparum-infected erythrocytes sequester in the placenta and attract activated mononuclear cells that secrete pro-inflammatory cytokines. Increased inflammatory cytokine levels in other microbial infections are associated with PTDs. To determine if such is the case in women with placental malaria, concentrations of interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha), interleukin-4 (IL-4), and IL-10 were measured in placental plasma of 391 malaria-infected and -uninfected Cameroonian women with premature and full-term deliveries. Risk factors for malaria-associated PTDs included peripheral and placental parasitemias greater than 1%, maternal anemia, elevated IL-10 levels, and low TNF-alpha:IL-10 ratios due to over-expression of IL-10. Alterations in cytokine levels may contribute to PTDs through the induction of anemia and/or altering cellular immune responses required for eliminating placental parasites.


Subject(s)
Cytokines/immunology , Malaria, Falciparum/immunology , Obstetric Labor, Premature , Placenta/immunology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Adult , Animals , Cameroon , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Malaria, Falciparum/parasitology , Pregnancy , Pregnancy Complications, Parasitic/parasitology
20.
Am J Trop Med Hyg ; 67(6): 566-70, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12518845

ABSTRACT

Between 1995 and 1998, a longitudinal study was conducted at 2 health centers in Cameroon to determine the prevalence of Plasmodium falciparum infection in pregnant women. There were 520 pregnant women enrolled at a rural clinic in the village of Etoa and 199 enrolled at the Biyem Assi Clinic in the city of Yaounde. In Etoa, pregnant women were younger, fewer took prophylaxis, and malaria prevalence was higher compared with pregnant women in the Biyem Assi Clinic. The prevalence of malaria infection peaked during the fourth month of pregnancy at both sites. Age was identified as a major risk factor because women < or = 20 years old were 1.8 and 3.4 times more likely to have malaria infection than women > 20 years old at Etoa and the Biyem Assi Clinic after adjusting for gravidae and other factors. The use of chemoprophylaxis and seasonality did not have a major effect.


Subject(s)
Malaria, Falciparum/epidemiology , Plasmodium falciparum/isolation & purification , Pregnancy Complications, Parasitic/epidemiology , Adolescent , Adult , Animals , Cameroon/epidemiology , Female , Humans , Longitudinal Studies , Malaria, Falciparum/parasitology , Parasitemia/epidemiology , Parasitemia/parasitology , Pregnancy , Pregnancy Complications, Parasitic/parasitology , Prevalence , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...