Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int Endod J ; 54(4): 585-600, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33210765

ABSTRACT

AIM: To model in vitro the contact between adult dental pulp stem cells (DPSCs) and lipoteichoic acid (LTA), a cell wall component expressed at the surface of most Gram-positive bacteria. METHODOLOGY: Human DPSCs obtained from impacted third molars were cultured and exposed to various concentrations of S. aureus LTA (0.1, 1.0 and 10 µg mL-1 ). The effects of LTA on DPSCs proliferation and apoptosis were investigated by MTT assay and flow cytometry. Mineralization of DPSCs was evaluated by alizarin red staining assay. Migration was investigated by microphotographs of wound-healing and Transwell migration assays. Reverse transcription polymerase chain reaction was used to examine the effects of LTA on p65 NF-κB translocation and TLR1, TLR2 or TLR6 regulation. Enzyme-linked immunosorbent assay was used to investigate LTA-stimulated DPSCs cytokine production. One-way or two-way ANOVA and Tukey post hoc multiple comparison were used for statistical analysis. RESULTS: DPSCs expressed TLR1, TLR2 and TLR6 involved in the recognition of various forms of LTA or lipoproteins. Exposure to LTA did not up- or down-regulate the mRNAs of TLR1, TLR2 or TLR6 whilst LPS acted as a potent inducer of them [TLR1 (P ≤ 0.05), TLR2 (P ≤ 0.001) and TLR6 (P ≤ 0.001)]. Translocation of p65 NF-κB to the nucleus was detected in LTA-stimulated cells, but to a lesser extent than LPS-stimulated DPSCs (P ≤ 0.001). The viability of cells exposed to LTA was greater than unstimulated cells, which was attributed to an increased proliferation and not to less cell death [LTA 1 µg mL-1 (P ≤ 0.001) and 10 µg mL-1 (P ≤ 0.01)]. For specific doses of LTA (1.0 µg mL-1 ), adhesion of DPSCs to collagen matrix was disturbed (P ≤ 0.05) and cells enhanced their horizontal mobility (P ≤ 0.001). LTA-stimulated DPSCs released IL-6 and IL-8 in a dose-dependent manner (P ≤ 0.0001). At all concentrations investigated, LTA did not influence osteogenic/odontoblastic differentiation. CONCLUSIONS: Human DPSCs were able to sense the wall components of Gram-positive bacteria likely through TLR2 signalling. Consequently, cells modestly proliferated, increased their migratory behaviour and contributed significantly to the local inflammatory response through cytokine release.


Subject(s)
Lipopolysaccharides , Osteogenesis , Adult , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines , Dental Pulp , Humans , Lipopolysaccharides/pharmacology , Staphylococcus aureus , Stem Cells , Teichoic Acids
2.
Parasite Immunol ; 36(1): 43-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24102464

ABSTRACT

Early interferon-gamma (IFN-γ) release by innate cells is critical to direct type 1 immune response able to control intracellular pathogens like Trypanosoma cruzi. Although CD56(bright) natural killer (NK) cells are reported to be potent early IFN-γ producers, other CD56(+) cells like CD56(dim) NK cells and NK-like T cells have recently been shown to also release IFN-γ. We have here studied the contribution of each CD56(+) lymphocyte populations in early IFN-γ production in both adults and neonates. On this purpose, we analysed the kinetics of IFN-γ production by RT-PCR, ELISA and flow cytometry from 2 h onwards after T. cruzi and IL-15 stimulation and sought for the responding CD56(+) cells. CD56(bright) and CD56(dim) CD16(-) NK cells were the more potent IFN-γ early producers in response to IL-15 and parasites in adults and neonates. In both age groups, the majority of IFN-γ producing cells were NK cells. However, on the contrary to neonates, CD3(+) CD56(+) NK-like T cells and CD3(+) CD56(-) 'classical' T cells also contributed to early IFN-γ production in adults. Altogether, our results support that whereas NK cells responded almost similarly in neonates and adults, cord blood innate CD56(+) and CD56(-) T cells displayed major quantitative and qualitative defects that could contribute to the well-known neonatal immune immaturity.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-15/immunology , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , Trypanosoma cruzi/immunology , Adult , CD56 Antigen/analysis , Chagas Disease/immunology , Chagas Disease/parasitology , Fetal Blood/immunology , Flow Cytometry , Humans , Infant, Newborn , Interferon-gamma/genetics , Killer Cells, Natural/metabolism , Kinetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocyte Subsets/metabolism
3.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 101-104, 2005. graf
Article in Spanish | LILACS | ID: lil-444165

ABSTRACT

The mechanisms of congenital transmission of Chagas disease remain largely unknown. To better understand the role of maternal immunology during pregnancy in congenital Chagas transmission, we studied the cytokine production and the parasitic load in three groups of mothers: infected mothers who transmitted the disease to their babies (M+B+-), infected mothers who did not transmit the disease to their babies (M+B-) and not infected mothers as a control group (M-B-). M+B+ mothers produced less IFNgamma and more IL-10 than the M+B- mothers, and they are not able to produce IL-2. M+B+ mothers showed a higher parasitic load. These results, indicated that the congenital Chagas transmission is associated with an immunological imbalance and a high parasitic load in the M+B+ mothers.


Subject(s)
Animals , Female , Humans , Pregnancy , Cytokines/biosynthesis , Pregnancy Complications, Infectious/immunology , Chagas Disease/immunology , Chagas Disease/transmission , Infectious Disease Transmission, Vertical , Trypanosoma cruzi/physiology , Cytokines/immunology , Chagas Disease/parasitology , Immunity, Cellular , Interferon-gamma/biosynthesis , Interferons/biosynthesis , Carrier State/immunology
4.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 96-100, 2005. graf
Article in Spanish | LILACS | ID: lil-444166

ABSTRACT

We have investigated if maternal T. cruzi infection could induce in utero innate and/or adaptive immune responses in uninfected neonates by measuring specific IgM and IgA antibodies in cord blood plasma, and by performing phenotypic and functional studies of umbilical cord blood cells of their newborns (M+B- group). We detected T. cruzi-specific IgM and IgA antibodies in M+B- cord blood, indicating they had mounted in utero a strong B cell response, although they are not infected. On the other hand, circulating T cells of such uninfected neonates displayed a low level of activation, as seen bya slightly increased expression of the activation markers CD45RO on CD4+ T cells and HLA-DR on CD8+ T cells, although the proportion of CD4+ and CD8+ T cells was unmodified as compared to newborns from uninfected mothers (MB- group). This activation did not give rise to a proliferative response upon stimulation by T. cruzi antigens in vitro. However, M+B- cells produced low levels of lymphokines (IFN-gamma and IL-13) upon mitogenic stimulation, which was not the case of M-B- newborn cells. Beside this, M+B- blood cells produced higher levels of inflammatory cytokines (IL-1b, IL-6, TNF-alpha) than M-B- cells when stimulated with the T. cruzi lysate or LPS, suggesting the over-activation of the innate response in M+B- newborns. Monocytes participated in such inflammatory response since M+B- purified cord blood monocytes produced higher levels of TNF- when incubated with LPS or a T. cruzi lysate than M-B- cells. Altogether, these results show that, even in the absence of congenital infection, maternal T. cruzi infection triggers in utero both adaptive and innate immune responses in their babies. This indicates that parasite circulating antigens have been transferred from mothers to their fetuses.


Subject(s)
Animals , Female , Humans , Infant, Newborn , Pregnancy , Chagas Disease/immunology , Chagas Disease/transmission , Infectious Disease Transmission, Vertical , Immunity, Maternally-Acquired/immunology , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Fetal Blood/immunology , Cytokines/biosynthesis , Pregnancy Complications, Parasitic/diagnosis , Chagas Disease/congenital , Immunity, Cellular , Immunoglobulin A , Immunoglobulin M
5.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 77-83, 2005. tab
Article in Spanish | LILACS | ID: lil-444170

ABSTRACT

Congenital transmission of T. cruzi in Cochabamba affects 6% of newborns from infected mothers. Only limited information is available on the type of transmitted parasites. However, it is well established that T. cruzi isolated from various vectors as well from host animals are highly heterogeneous. In our presentation we analyse aspects of molecular heterogeneity of T. cruzi and we review methods used for the molecular typing of T. cruzi lineages. Experimentally, we performed the PCR amplification of [quot ]Sequence-characterised region Markers[quot ] for typing T. cruzi isolated from umbilical blood of newborns in Cochabamba. We compared these results with those we obtained from general infected population. All 16 analysed, congenitally infected samples were of lineage IId. Our data also indicated that this lineage was found in about 80% of samples originated from general infected population in Cochabamba.


Subject(s)
Animals , Humans , Chagas Disease/congenital , Genetic Heterogeneity , Trypanosoma cruzi/genetics , DNA, Protozoan/analysis , Chagas Disease/diagnosis , Polymerase Chain Reaction/methods , Trypanosoma cruzi/classification , Trypanosoma cruzi/isolation & purification
6.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 73-76, 2005. tab
Article in Spanish | LILACS | ID: lil-444171

ABSTRACT

In the endemic regions of Bolivia the infection of the feminine population in fertile age by T. cruzi is frequent (20 to 50 % of the women in fertile age) and the rate of fetal maternal transmission is of approximately 5%. A great percentage of infected women do not transmit the infection to the fetus. The intention of the present study carried out at the Maternal-Infantile Hospital Germán Urquidi of Cochabamba (Bolivia) is to contribute to the knowledge regarding the pregnancy and birth of a newborn of Chagas infected women who do not transmit the infection to the fetus. 2124 mothers and 2,155 newborns were studied. The prevalence of infection by T. cruzi among these pregnant women is of 26,3%. Two groups of mothers were studied: 554 that presented infection by T. cruzi (group M+B-) and 1520 not infected (group control M-B-). Both groups of mothers are comparable in their anthropometric and obstetrical antecedents. The mothers (M+B+) are in average older than those not infected (p<0.05), which will probably have an influence on the number of gestations and abortion antecedents, which were of p<0.05 and p=0.01 respectively. Among the different anthropometric and biological parameters studied in newborns of groups M+B- and M-B -, no statistically significant differences between both groups were found. It can be inferred that the chronic maternal infection by T. cruzi seems to have no clinical influence, neither on the course of the pregnancy nor during birth, if a group of T. cruzi infected mothers is compared to a non infected group.


Subject(s)
Female , Humans , Infant, Newborn , Pregnancy , Adult , Pregnancy Complications, Parasitic/epidemiology , Chagas Disease/epidemiology , Infectious Disease Transmission, Vertical , Pregnancy Outcome , Anthropometry , Apgar Score , Bolivia/epidemiology , Case-Control Studies , Chagas Disease/diagnosis , Chagas Disease/transmission , Prevalence
7.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 68-72, 2005. graf, tab
Article in Spanish | LILACS | ID: lil-444172

ABSTRACT

Pathogens may impair reproduction in association or not with congenital infections. We have investigated the effect of acute infection with Trypanosoma cruzi, the protozoan agent of Chagas disease, on reproduction of female mice. In the acute, parasitemic, phase of the infection, female mice were totally unable to reproduce. Most of them (80%) were infertiles and did not develop any gestation. In the few gravid infected mice, implantation numbers were as in uninfected control mice. However, their fetuses presented a weight meanly reduced by 40% as compared to those of uninfected females, and all of them died during the gestation or whithin 48 h after birth. Such massive mortality did not result from congenital infection, which did not occur. The infertility and the fetal mortality occuring early in gestation (resorptions) were significantly correlated with a high maternal parasitemia, whereas later fetal mortality was associated with the presence of intracellular parasites in the utero-placental unit. The decidua was particularly receptive to T. cruzi multiplication, since this tissue harboured 125 fold more amastigotes than the maternal heart or other placental tissues. In addition, placentas of dead fetuses presented histopathological lesions (inflammatory infiltrates, fibrine deposits and ischemic necrosis). Such harmfull effects of acute infection were not observed when female mice were in the chronic phase of the infection, since these reproduce normally. Their fetuses only suffered from moderate and reversible growth retardation. These results indicate that, following the maternal parasite burden, T. cruzi infection may induce very deleterious effects on gestation.


Subject(s)
Animals , Female , Pregnancy , Chagas Disease/complications , Infertility/parasitology , Fetal Death/parasitology , Pregnancy Complications, Parasitic , Trypanosoma cruzi , Acute Disease , Chronic Disease , Mice , Mice, Inbred BALB C , Fetal Death/pathology , Necrosis , Placenta/parasitology , Trypanosoma cruzi/pathogenicity
8.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 65-67, 2005. ilus, tab
Article in Spanish | LILACS | ID: lil-444173

ABSTRACT

PCR is a potentially interesting diagnostic tool to detect congenital T. cruzi infection. We have compared the sensitivity and capacity of a battery of T. cruzi PCR primers to detect the complete spectrum of known T. cruzi lineages, in order to improve and simplify the detection of infection in neonatal blood. We found that the primers Tcz1/Tcz2, targeting the 195 bp satellite repeat, detected all the parasitic lineages with the same sensitivity For all other tested primers (nDNA primers: BP1/BP2, 01/02, Pon1/ Pon2 and Tca1/Tca2; kDNA primers: S35VS36, 121/122), either, the intensity of amplicons varied according to T. cruzi lineages, or the assess were less sensitive. In order to better assess such PCR protocol, we assayed 311 samples of neonatal blood previously tested with parasitological methods. Reliability of our PCR test was demonstrated since all the 18 blood samples from newborns with congenital T. cruzi infection were positive, whereas the remaining samples (30 from control newborns of uninfected mothers and 262 out of 263 from babies, parasitologically negative, born from infected mothers) were negative. As our PCR method is simple, reliable, robust and cheap, it appears suitable for the detection of T. cruzi infection in neonatal blood.


Subject(s)
Animals , Humans , Infant, Newborn , Chagas Disease/congenital , Chagas Disease/diagnosis , Polymerase Chain Reaction/standards , Trypanosoma cruzi/isolation & purification , DNA, Protozoan/blood , Infectious Disease Transmission, Vertical , DNA Primers , Reproducibility of Results , Sensitivity and Specificity , Fetal Blood/parasitology , Trypanosoma cruzi/genetics
9.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 62-64, 2005. graf, tab
Article in Spanish | LILACS | ID: lil-444174

ABSTRACT

This study compares the levels of specific antibodies IgM and IgA for Chagas in samples of blood from newborns. Three groups of cord blood samples have been analysed: a group of 42 samples from newborns, displaying positive parasitemia, of seropositive mothers (M+B+), 68 samples from newborns with negative parasitemia whose mothers were seropositive (M+B-) and a group of 45 control newborns coming from mothers with negative serology for Chagas. From the 42 M+B+ samples with congenital Chagas disease, 81 and 82.9% displayed detectable levels of IgM and IgA antibodies, respectively In the M+B- group, 70.6 and 33.8% presented antibodies of IgM and IgA classes, respectively, whereas in the control group M-B-, we detected 6% and 11.1% of IgM and IgA antibodies, respectively. The calculated sensitivity of detection of congenital cases using IgM or IgA antibodies was of 82.9% and 80.9% respectively, whereas the specificity of detection was of 29.4% for IgM antibodies and of 66.1% for IgA antibodies.


Subject(s)
Animals , Humans , Infant, Newborn , Chagas Disease/congenital , Chagas Disease/diagnosis , Immunoglobulin A/blood , Immunoglobulin M/blood , Trypanosoma cruzi/immunology , Case-Control Studies , Chagas Disease/immunology , Enzyme-Linked Immunosorbent Assay , Sensitivity and Specificity
10.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 58-61, 2005. graf, tab, ilus
Article in Spanish | LILACS | ID: lil-444175

ABSTRACT

The aim of this study was to validate the method of microhematocrit tube, as a rapid method to estimate the parasitemia in blood and to associate the parasites concentration with the morbidity and mortality of new born children with congenital Chagas diseases. Our results were determined experimentally and shown that the detection limit of the microhematocrit tube method is 40 parasites/ml when at least one of the four observed tubes is positive. Besides, it was also established that when the four examined tubes are positive the parasitemia in blood reaches more than 100 parasites/ml. It is important to highlight the modification made by our laboratory in the microscopic observation of the microhematocrit tubes with respect to the methodology used by previous investigators. A positive association exists between a high number of parasites in blood and the morbi-mortality of the newly born children with congenital chagas. The results of positive association between the parasitic load and the morbility and mortality could constitute an argument to understand the possible role of the parasite in the pathology of the disease.


Subject(s)
Humans , Animals , Male , Female , Infant, Newborn , Mice , Parasite Egg Count/methods , Chagas Disease/congenital , Chagas Disease/parasitology , Parasitemia , Trypanosoma cruzi/isolation & purification , Birth Weight , Bolivia/epidemiology , Chagas Disease/diagnosis , Hematocrit/instrumentation , Hematocrit/methods , Parasitemia/mortality , Sensitivity and Specificity , Umbilical Cord
11.
Rev. Soc. Bras. Med. Trop ; 38(supl.2): 17-20, 2005. tab
Article in Spanish | LILACS | ID: lil-444185

ABSTRACT

In Bolivia, the prevalence of infection by T. cruzi in women in fertile age can vary between 20 and 60%. The present study made in the Maternity Germin Urquidi of Cochabamba - Bolivia, it has demonstrated, that 19.9% of the mothers who go to this hospitable center to be taken care of in the childbirth, they are carrying of the infection and that 4,6% of them, they are going to transmit, by transplacentaria route, the infection to its babies. Of the 71 children born with congenital Chagas, only 47,8 % present/display some type of alteration or of development(Apgar to 1 minute low, BPN, prematuridad, pathological dismadurez) or signs (SDR, hepatomegalia, esplenomegalia, neurological signs, cardiomegalia, anasarca, petequias). When investigating the effect of the differences in the vectorial density (low, medium and high) of the zone of maternal residence, on the transmission of the infection of the mother infected to the fetus, we concluded that the rate of transmission of the congenital infection of T. cruzi is not modified by the level of endemicidad of the zone of maternal residence. By another infected new born sides whose mothers reside in zones of high endemicidad present/display, most frequently and of significant way, Apgar to 1 minute < to 7, low weight when being born and prematuridad or an association of these alterations with respiratory syndrome of distress or anasarca, when one compares them with new born of resident mothers in the zones of loss or medium endemicidad, mortality in this group is greater. These results suggest calls to account it of the mothers, in areas of high endemicidad, she is associate with a serious increase in the risk of Disease of newborn severe and mortal congenital Chagas in.


Subject(s)
Humans , Animals , Male , Female , Pregnancy , Infant, Newborn , Adult , Chagas Disease/congenital , Endemic Diseases , Infectious Disease Transmission, Vertical , Insect Vectors/physiology , Pregnancy Complications, Parasitic , Apgar Score , Bolivia/epidemiology , Demography , Chagas Disease/epidemiology , Chagas Disease/transmission , Epidemiologic Factors , Population Density , Prevalence , Trypanosoma cruzi/physiology
12.
Vaccine ; 22(15-16): 1868-72, 2004 May 07.
Article in English | MEDLINE | ID: mdl-15121297

ABSTRACT

We have developed an experimental model of vaccination against the infection with the protozoa Trypanosoma cruzi, the agent of Chagas disease in Latin America. Vaccination was performed with Trypanosoma rangeli, a non-pathogenic protozoa sharing many antigens with T. cruzi. It strongly protected BALB/c mice, sharply reducing parasitaemia and mortality rate of the acute T. cruzi infection. The aim of the present work was to complete our previous study on the production of IFN-gamma and IL-10 in this vaccination model by investigating the production of IL-12p35 and p40, IL-18, TNF, TNF soluble receptors (sTNFR), and nitric oxide (NO), factors known to play a key role in the outcome of T. cruzi infection. We show that the protection obtained against the acute T. cruzi infection was surprisingly associated with reduced circulating levels of IL-18 and NO, whereas the release of IL-12p40 was enhanced in comparison to non-vaccinated infected animals. IL-12p35 remained undetectable in infected animals, vaccinated or not. The balance between sTNFR and TNF suggested a decrease of TNF bioactivity in vaccinated mice. These results show that the protection induced by the vaccination with T. rangeli against a challenging infection with T. cruzi is not associated with the strong type 1 immune response usually involved in the control of intracellular pathogens, particularly questioning the protective role of NO during the acute phase of T. cruzi infection.


Subject(s)
Chagas Disease/immunology , Chagas Disease/prevention & control , Cytokines/biosynthesis , Nitric Oxide/biosynthesis , Protozoan Vaccines/immunology , Receptors, Tumor Necrosis Factor/biosynthesis , Trypanosoma cruzi/immunology , Trypanosoma/immunology , Animals , Interleukin-12/biosynthesis , Interleukin-18/biosynthesis , Mice , Mice, Inbred BALB C , Survival Analysis , Tumor Necrosis Factor-alpha/biosynthesis , Vaccination
13.
Am J Reprod Immunol ; 49(2): 101-12, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12765349

ABSTRACT

PROBLEM: To determine if interleukin-16 (IL-16), IL-17, and IL-18 are present at the murine fetomaternal interface during pregnancy as a first step towards investigating their roles in fetomaternal relationship. METHODS: Expression of IL-16, IL-17, and IL-18, was assessed by immunohistochemistry (IHC) in the BALB/c x BALB/k (H2d x H2k), and the CBA/J x BALB/c non-abortion prone, and CBA/J x DBA/2 abortion prone matings. Enzyme-linked immunosorbent assay (ELISA) were performed for the two latter cytokines to compare local production in the abortion prone CBA/J x DBA/2 versus the non-abortion prone CBA/J x BALB/c matings. RESULTS: Expression of IL-17 was borderline. The anti-IL-16 staining specifically localized in the uterine stroma and glandular epithelium and was rather low in the placenta. IL-18 staining started in the peri-implantation uterus in the basal proliferative stroma, and was also traced, although weaker, in the glandular epithelium. In the immediate post-implantation period, a weak stromal staining persisted but there was a strong labeling of the ectoplacental cone. Interestingly, when the ectoplacental cone differentiates into placenta having a major histocompatibility complex (MHC) class I + spongiotrophoblast and a (MHC class I-) labyrinth, a very strong transient labeling of uterine natural killer (u-NK) cells was found. Later in gestation, IL-18 was also produced by giant cell and spongiotrophoblast. Finally, we compared by ELISA the production of IL-17/-18 in CBA/J x DBA/2 and CBA/J x BALB/c matings. We detected significantly more IL-18 in the non-abortion prone combination decidua or placenta. CONCLUSION: The three cytokines IL-16, IL-17, and IL-18 were detected at the fetomaternal interface with a tissue specific, stage-dependent distribution. The predominance of IL-18 secretion in the non-resorption prone matings lead us to question the general validity of the classical T-helper (Th)1/2 paradigm.


Subject(s)
Interleukin-16/metabolism , Interleukin-17/metabolism , Interleukin-18/metabolism , Placenta/metabolism , Animals , Decidua/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Interleukin-16/analysis , Interleukin-17/analysis , Interleukin-18/analysis , Mice , Pregnancy , Time Factors
14.
Infect Immun ; 68(9): 5430-4, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10948177

ABSTRACT

The possibility of maternal in utero modulation of the innate and/or adaptive immune responses of uninfected newborns from Trypanosoma cruzi-infected mothers was investigated by studying the capacity of their whole blood cells to produce cytokines in response to T. cruzi lysate or lipopolysaccharide-plus-phytohemagglutinin (LPS-PHA) stimulation. Cells of such newborns occasionally released gamma interferon (IFN-gamma) and no interleukin-2 (IL-2) and IL-4 upon specific stimulation, while their mothers responded by the production of IFN-gamma, IL-2, and IL-4. Infection in mothers was also associated with a hyperactivation of maternal cells and also, strikingly, of cells of their uninfected neonates, since their release of proinflammatory (IL-1beta, IL-6, and tumor necrosis factor alpha [TNF-alpha]) as well as of anti-inflammatory (IL-10 and soluble TNF receptor) cytokines or factors was upregulated in the presence of LPS-PHA and/or parasite lysate. These results show that T. cruzi infection in mothers induces profound perturbations in the cytokine response of their uninfected neonates. Such maternal influence on neonatal innate immunity might contribute to limit the occurrence and severity of congenital infection.


Subject(s)
Chagas Disease/immunology , Cytokines/biosynthesis , Pregnancy Complications, Parasitic/immunology , Female , Humans , Immunity, Maternally-Acquired , Infant, Newborn , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-4/biosynthesis , Interleukin-6/biosynthesis , Pregnancy , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
15.
Int Immunol ; 12(2): 223-30, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10653858

ABSTRACT

After infection with some viruses and intracellular parasites, antibody production is restricted to IgG2a. We first observed that, whereas live viruses such as lactate dehydrogenase-elevating virus (LDV) or mouse adenovirus induced mostly an IgG2a response, a large proportion of antibodies produced against killed viruses were IgG1. This IgG1 antiviral response was suppressed when live virions were added to inactivated viral particles. These results indicate that the IgG2a preponderance is related to the infectious process itself rather than to the type of antigen involved. Since IFN-gamma is known to stimulate IgG2a production by activated B lymphocytes and to be secreted after infection, we examined the role of this cytokine in the antibody isotypic distribution caused by LDV. Most IgG2a responses were relatively unaffected in mice deficient for the IFN-gamma receptor or treated with anti-IFN-gamma antibody. A similar IFN-gamma-independent IgG2a secretion was observed after infection with the parasites Toxoplasma gondii and Trypanosoma cruzi. However, the IFN-gamma-independent IgG2a production triggered by infection still required the presence of functional T(h) lymphocytes. Therefore, signal(s) other than IFN-gamma secretion may explain the T(h)-dependent isotypic bias in antibody secretion triggered by viruses and parasites.


Subject(s)
Immunoglobulin G/biosynthesis , Interferon-gamma/pharmacology , Protozoan Infections/immunology , Virus Diseases/immunology , Adenoviridae/immunology , Adenoviridae Infections/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/blood , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Arterivirus Infections/immunology , Chagas Disease/immunology , Female , Immunoglobulin G/blood , Lactate dehydrogenase-elevating virus/immunology , Mice , Mice, Inbred CBA , Spleen/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Trypanosoma cruzi
16.
Infect Immun ; 67(11): 5579-86, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10531203

ABSTRACT

To better understand the role of tumor necrosis factor (TNF) during Trypanosoma cruzi infection in BALB/c mice, we have investigated the kinetics of circulating tumor necrosis factor (TNF), soluble TNF receptor 1 (sTNR1), and sTNFR2 levels, as well as the interactions between such factors, in relation to parasitemia, cachexia, and mortality of acutely infected animals. Our data show that the parasitemic phase of T. cruzi infection in mice is associated with high levels of circulating TNF and sTNFR2, resulting in the formation of cytokine-receptor complexes and some degree of neutralization of TNF bioactivity. Although sTNR2 levels always exceeded TNF levels, low sTNFR/TNF circulating ratios were associated with cachexia in all infected mice, whereas the lowest ratios were observed in dying animals harboring the highest parasitemia. We also studied the modulation of sTNFR/TNF ratios induced by anti-TNF antibodies administered to infected animals and their consequences on the outcome of the infection. The injection of anti-TNF monoclonal antibody (MAb) TN3 into infected mice resulted in a paradoxical overproduction of TNF (associated with a higher parasitemia), lowered the sTNFR/TNF circulating ratios, and considerably worsened cachexia and mortality of animals. Another anti-TNF MAb (1F3F3) decreased the in vivo availability of TNF as well as parasite levels and reduced cachexia. Altogether, such results highlight that, besides playing a beneficial role early in infection, TNF also triggers harmful effects in the parasitemic phase, which are limited by the in vivo simultaneous endogenous production of soluble receptors.


Subject(s)
Cachexia/etiology , Chagas Disease/mortality , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Acute Disease , Animals , Antibodies, Monoclonal/therapeutic use , Cachexia/therapy , Chagas Disease/complications , Male , Mice , Mice, Inbred BALB C , Receptors, Tumor Necrosis Factor/blood , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/immunology
17.
Clin Exp Immunol ; 113(1): 59-64, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9697984

ABSTRACT

We examined the effects of IL-10 on tumour necrosis factor-alpha (TNF-alpha) and NO production by LPS-activated macrophages and on the ability of these cells to control Trypanosoma cruzi infection. We first observed that the addition of rIL-10 to macrophages of the J774 cell line decreased their synthesis of TNF-alpha but increased their release of NO in a dose-dependent manner. In parallel, treatment of J774 cells with rIL-10 resulted in a better control of T. cruzi infection involving up-regulation of NO synthesis, as it was not observed in presence of N-nitro-L-arginine methyl ester (L-NAME), a competitive inhibitor of NO synthase. The enhancing effect of rIL-10 on NO production was not observed on peritoneal macrophages from wild-type C57Bl/6 mice, but well on macrophages from IL-10 knock-out mice. The control of NO production by endogenous IL-10 was confirmed by the demonstration that neutralization of IL-10 secreted by LPS-activated macrophages from wild-type mice inhibited their production of NO and, in parallel, their ability to control T. cruzi infection. Taken together, these data demonstrate that both exogenous and endogenous IL-10 up-regulate the production of NO by LPS-activated macrophages and improve thereby their ability to clear T. cruzi infection.


Subject(s)
Chagas Disease/prevention & control , Interleukin-10/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Nitric Oxide/biosynthesis , Up-Regulation , Animals , Chagas Disease/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Trypanosoma cruzi
18.
Parasite Immunol ; 17(11): 561-8, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8817602

ABSTRACT

BALB/c male mice acutely infected with Trypanosoma cruzi underwent a severe weight loss (around 20%, from day 18 to 31 post-infection), when compared to age-matched uninfected animals. Though mice regained weight later, when blood parasites were hardly detectable, wasting extended over the chronic phase of infection. The onset and the magnitude of weight loss were related to the mouse susceptibility to infection, since they were respectively earlier and higher in male mice which will die than in surviving ones, in males than in females, and in BALB/c than in B6D2 [(C57B1/6 x DBA/2)F1], a mouse strain more resistant to infection. Fat weight of infected mice (male BALB/c) was reduced by 60 to 80%, whereas lean mass was unaffected and water content rose by 6 to 10% in acute and chronic infection. Haematocrit was also decreased by 15-16% in acute infection. Animals failed to compensate their energetic loss since their food intake remained similar to that of uninfected animals. Injections of neutralizing anti-TNF-alpha monoclonal antibody into infected male mice, during the first two weeks but not later in infection, significantly attenuated the weight loss. Early administration of anti-IL-6 or anti-IFN-gamma MoAbs did not improve the mouse wasting. Taken together, these data show that TNF is a key agent of cachexia occurring in the acute T. cruzi infection in mice.


Subject(s)
Cachexia , Chagas Disease/physiopathology , Trypanosoma cruzi , Animals , Body Composition , Body Weight , Chagas Disease/immunology , Eating , Female , Hematocrit , Interferon-gamma/immunology , Interleukin-6/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Tumor Necrosis Factor-alpha/immunology
19.
Exp Parasitol ; 80(3): 499-506, 1995 May.
Article in English | MEDLINE | ID: mdl-7729485

ABSTRACT

The levels of fibronectin (FN), a multifunctional glycoprotein known to mediate in vitro Trypanosoma cruzi-host cell adhesion, were measured in the plasma of T. cruzi-infected BALB/c mice. The infection induced a long-lasting increase of fibronectin levels during the acute parasitemic phase of the disease. Immunoblotting analysis showed the occurrence of lower-molecular-size FN fragments in the plasma of acutely infected animals, suggesting an infection-related FN degradation. FN levels were found to be significantly lower in dying mice harboring higher parasitemias than in surviving animals. A weak level of natural IgM against the RGD adhesion site of FN was detected before and during the first 3 weeks of infection. The level was significantly higher in surviving mice. From the fourth week postinfection, a significant increase in the levels of anti-RGD antibodies coincided with a decrease of circulating FN. These antibodies were mainly of the IgM, IgG1, and IgG2a isotypes. Taken together, these data suggest that both FN and anti-FN antibodies may contribute to the outcome of T. cruzi infection in mice.


Subject(s)
Antibodies, Helminth/blood , Chagas Disease/blood , Fibronectins/blood , Oligopeptides/immunology , Parasitemia/blood , Acute Disease , Amino Acid Sequence , Analysis of Variance , Animals , Blotting, Western , Cell Adhesion , Chagas Disease/immunology , Fibronectins/chemistry , Fibronectins/immunology , Immunoglobulins/blood , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oligopeptides/chemistry , Parasitemia/immunology , Receptors, Immunologic/chemistry , Receptors, Immunologic/immunology
20.
Parasitol Today ; 11(3): 94-9, 1995 Mar.
Article in English | MEDLINE | ID: mdl-15275360

ABSTRACT

Immunoglobulins, parasite circulating antigens, immune cells, cytokines and other cell-related products can be transferred from infected mothers to their young. They can combine their effects to interact with the invading parasites, as well as to induce a long-term modulation of the offspring's capacity to mount an immune response to subsequent exposure to parasites. The protective effect of maternally derived antibodies may be limited by the selective transfer of immunoglobulin isotypes. Maternal antibodies may also prevent the priming of specific cells in offspring or inhibit the progeny's antibody production by interacting with B-cell receptors or with the idiotypic repertoire. The potentially beneficial priming effect of transferred parasitic antigens may be altered by the Th2-cell-biased foetal environment and such antigens may also induce deletion or anergy of T- and B-cell clones in offspring. Therefore, besides protective effects, maternal infection may downregulate the offspring's immune response. If such hyporesponsiveness may be clearly harmful (in increasing the risk or in worsening congenital or postnatally acquired infections in offspring), it can also be beneficial (in limiting the pathogenesis of some infections). Here, Yves Carlier and Carine Truyens review the rationale of these complex foeto-maternal relationships in parasitic diseases.

SELECTION OF CITATIONS
SEARCH DETAIL
...