1
|
Desale H, Buekens P, Alger J, Cafferata ML, Harville EW, Herrera C, Truyens C, Dumonteil E. Epigenetic signature of exposure to maternal Trypanosoma cruzi infection in cord blood cells from uninfected newborns. Epigenomics 2022; 14:913-927. [PMID: 36039408 PMCID: PMC9475499 DOI: 10.2217/epi-2022-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To assess the epigenetic effects of in utero exposure to maternal Trypanosoma cruzi infection. Methods: We performed an epigenome-wide association study to compare the DNA methylation patterns of umbilical cord blood cells from uninfected babies from chagasic and uninfected mothers. DNA methylation was measured using Infinium EPIC arrays. Results: We identified a differential DNA methylation signature of fetal exposure to maternal T. cruzi infection, in the absence of parasite transmission, with 12 differentially methylated sites in B cells and CD4+ T cells, including eight protein-coding genes. Conclusion: These genes participate in hematopoietic cell differentiation and the immune response and may be involved in immune disorders. They also have been associated with several developmental disorders and syndromes.
Collapse
Affiliation(s)
- Hans Desale
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| | - Pierre Buekens
- Department of Epidemiology, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA 70112, USA
| | - Jackeline Alger
- Instituto de Enfermedades Infecciosas y Parasitologia Antonio Vidal, Tegucigalpa, Honduras.,Ministry of Health, Hospital Escuela, Tegucigalpa, Honduras
| | - Maria Luisa Cafferata
- Unidad de Investigación Clínica y Epidemiológica Montevideo (UNICEM), Hospital de Clínicas, Montevideo, 11600, Uruguay
| | - Emily Wheeler Harville
- Department of Epidemiology, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA 70112, USA
| | - Claudia Herrera
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, & ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Dumonteil
- Department of Tropical Medicine, Tulane University School of Public Health & Tropical Medicine & Tulane University Vector-Borne & Infectious Disease Research Center, New Orleans, LA 70112, USA
| |
Collapse
|
2
|
Carlier Y, Truyens C, Muraille E. Is Antibody-Dependent Enhancement of Trypanosoma cruzi Infection Contributing to Congenital/Neonatal Chagas Disease? Front Immunol 2021; 12:723516. [PMID: 34566981 PMCID: PMC8461104 DOI: 10.3389/fimmu.2021.723516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/26/2021] [Indexed: 11/13/2022] Open
Abstract
The newborns of women infected with the parasite Trypanosoma cruzi (the agent of Chagas disease) can be infected either before birth (congenitally), or after birth (as e.g., by vector route). Congenital Chagas disease can induce high levels of neonatal morbidity and mortality. Parasite-infected pregnant women transmit antibodies to their fetus. Antibodies, by opsonizing parasites, can promote phagocytosis and killing of T. cruzi by cells expressing FcγR, on the mandatory condition that such cells are sufficiently activated in an inflammatory context. Antibody-dependent enhancement (ADE) is a mechanism well described in viral infections, by which antibodies enhance entry of infectious agents into host cells by exploiting the phagocytic FcγR pathway. Previously reported Chagas disease studies highlighted a severe reduction of the maternal-fetal/neonatal inflammatory context in parasite-transmitting pregnant women and their congenitally infected newborns. Otherwise, experimental observations brought to light ADE of T. cruzi infection (involving FcγR) in mouse pups displaying maternally transferred antibodies, out of an inflammatory context. Herein, based on such data, we discuss the previously unconsidered possibility of a role of ADE in the trans-placental parasite transmission, and/or the development of severe and mortal clinical forms of congenital/neonatal Chagas disease in newborns of T. cruzi-infected mothers.
Collapse
Affiliation(s)
- Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium.,Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Carine Truyens
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Eric Muraille
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| |
Collapse
|
3
|
Kemmerling U, Osuna A, Schijman AG, Truyens C. Congenital Transmission of Trypanosoma cruzi: A Review About the Interactions Between the Parasite, the Placenta, the Maternal and the Fetal/Neonatal Immune Responses. Front Microbiol 2019; 10:1854. [PMID: 31474955 PMCID: PMC6702454 DOI: 10.3389/fmicb.2019.01854] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Chagas disease (CD), caused by the protozoan parasite Trypanosoma cruzi, is considered a neglected tropical disease by the World Health Organization. Congenital transmission of CD is an increasingly relevant public health problem. It progressively becomes the main transmission route over others and can occur in both endemic and non-endemic countries. Though most congenitally infected newborns are asymptomatic at birth, they display higher frequencies of prematurity, low birth weight, and lower Apgar scores compared to uninfected ones, and some suffer from severe symptoms. If not diagnosed and treated, infected newborns are at risk of developing disabling and life-threatening chronic pathologies later in life. The success or failure of congenital transmission depends on interactions between the parasite, the placenta, the mother, and the fetus. We review and discuss here the current knowledge about these parameters, including parasite virulence factors such as exovesicles, placental tropism, potential placental defense mechanisms, the placental transcriptome of infected women, gene polymorphism, and the maternal and fetal/neonatal immune responses, that might modulate the risk of T. cruzi congenital transmission.
Collapse
Affiliation(s)
- Ulrike Kemmerling
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Antonio Osuna
- Grupo de Bioquímica y Parasitología Molecular, Departamento de Parasitología, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - Alejandro Gabriel Schijman
- Molecular Biology of Chagas Disease Laboratory, Genetic Engineering and Molecular Biology Research Institute Dr. Héctor Torres (INGEBI-CONICET), Buenos Aires, Argentina
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
4
|
Carlier Y, Truyens C. Congenital Chagas disease as an ecological model of interactions between Trypanosoma cruzi parasites, pregnant women, placenta and fetuses. Acta Trop 2015; 151:103-15. [PMID: 26293886 DOI: 10.1016/j.actatropica.2015.07.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 12/31/2022]
Abstract
The aim of this paper is to discuss the main ecological interactions between the parasite Trypanosoma cruzi and its hosts, the mother and the fetus, leading to the transmission and development of congenital Chagas disease. One or several infecting strains of T. cruzi (with specific features) interact with: (i) the immune system of a pregnant woman whom responses depend on genetic and environmental factors, (ii) the placenta harboring its own defenses, and, finally, (iii) the fetal immune system displaying responses also susceptible to be modulated by maternal and environmental factors, as well as his own genetic background which is different from her mother. The severity of congenital Chagas disease depends on the magnitude of such final responses. The paper is mainly based on human data, but integrates also complementary observations obtained in experimental infections. It also focuses on important gaps in our knowledge of this congenital infection, such as the role of parasite diversity vs host genetic factors, as well as that of the maternal and placental microbiomes and the microbiome acquisition by infant in the control of infection. Investigations on these topics are needed in order to improve the programs aiming to diagnose, manage and control congenital Chagas disease.
Collapse
Affiliation(s)
- Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), CP 616, Route de Lennik 808, 1070 Bruxelles, Belgium; Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, Suite 2210, 1440 Canal Street, New Orleans, LA 70112-2797, USA.
| | - Carine Truyens
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), CP 616, Route de Lennik 808, 1070 Bruxelles, Belgium.
| |
Collapse
|
5
|
Cardoni RL, Antunez MI. Outcome ofTrypanosoma cruziinfection in pregnant BALB/c mice. ANNALS OF TROPICAL MEDICINE AND PARASITOLOGY 2013; 98:883-7. [PMID: 15667721 DOI: 10.1179/00034980x3234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- R L Cardoni
- Instituto Nacional de Parasitología Dr M. Fatala Chabén, ANLIS 'Dr C. G. Malbrán', Av. Paseo Colón 568, 1063 Buenos Aires, Argentina.
| | | |
Collapse
|
6
|
Cencig S, Coltel N, Truyens C, Carlier Y. Fertility, gestation outcome and parasite congenital transmissibility in mice infected with TcI, TcII and TcVI genotypes of Trypanosoma cruzi. PLoS Negl Trop Dis 2013; 7:e2271. [PMID: 23785533 PMCID: PMC3681732 DOI: 10.1371/journal.pntd.0002271] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 05/03/2013] [Indexed: 12/21/2022] Open
Abstract
This work aims to compare the effects of acute or chronic infections with the T. cruzi genotypes TcI (X10 strain), TcII (Y strain) and TcVI (Tulahuen strain) on fertility, gestation, pup growth and the possible vertical transmission of parasites in BALB/c mice. The occurrence of congenital infection was evaluated by microscopic examination of blood and/or qPCR on blood and heart in newborn pups and/or older offspring submitted to cyclophosphamide-induced immunosuppression in order to detect possible cryptic congenital infection. Altogether, the results show that: i) for the three strains tested, acute infection occurring after the embryo implantation in the uterus (parasite inoculation 4 days before mating), or close to delivery (parasite inoculation on day 13 of gestation), prevents or severely jeopardizes gestation outcome (inducing pup mortality and intra-uterine growth retardation); ii) for the three strains tested, gestation during chronic infection results in intra-uterine growth retardation, whereas re-inoculation of TcVI parasites during gestation in such chronically infected mice, in addition, strongly increases pup mortality; iii) congenital infection remains a rare consequence of infection (occurring in approximately 4% of living pups born to acutely infected dams); iv) PCR, detecting parasitic DNA and not living parasites, is not convenient to detect congenial infection close to delivery; v) transmission of parasites by breast milk is unlikely. This study should encourage further investigations using other parasite strains and genotypes to explore the role of virulence and other factors, as well as the mechanisms of such effects on gestation and on the establishment of congenital infection.
Collapse
Affiliation(s)
- Sabrina Cencig
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nicolas Coltel
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Carine Truyens
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Yves Carlier
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
7
|
Pérez AR, Berbert LR, Lepletier A, Revelli S, Bottasso O, Silva-Barbosa SD, Savino W. TNF-α is involved in the abnormal thymocyte migration during experimental Trypanosoma cruzi infection and favors the export of immature cells. PLoS One 2012; 7:e34360. [PMID: 22461911 PMCID: PMC3312912 DOI: 10.1371/journal.pone.0034360] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 02/27/2012] [Indexed: 11/28/2022] Open
Abstract
Previous studies revealed a significant production of inflammatory cytokines together with severe thymic atrophy and thymocyte migratory disturbances during experimental Chagas disease. Migratory activity of thymocytes and mature T cells seem to be finely tuned by cytokines, chemokines and extracellular matrix (ECM) components. Systemic TNF-α is enhanced during infection and appears to be crucial in the response against the parasite. However, it also seems to be involved in disease pathology, since it is implicated in the arrival of T cells to effector sites, including the myocardium. Herein, we analyzed the role of TNF-α in the migratory activity of thymocytes in Trypanosoma cruzi (T. cruzi) acutely-infected mice. We found increased expression and deposition of TNF-α in the thymus of infected animals compared to controls, accompanied by increased co-localization of fibronectin, a cell migration-related ECM molecule, whose contents in the thymus of infected mice is also augmented. In-vivo studies showed an enhanced export of thymocytes in T. cruzi-infected mice, as ascertained by intrathymic injection of FITC alone or in combination with TNF-α. The increase of immature CD4+CD8+ T cells in secondary lymphoid organs was even more clear-cut when TNF-α was co-injected with FITC. Ex-vivo transmigration assays also revealed higher number of migrating cells when TNF-α was added onto fibronectin lattices, with higher input of all thymocyte subsets, including immature CD4+CD8+. Infected animals also exhibit enhanced levels of expression of both mRNA TNF-α receptors in the CD4+CD8+ subpopulation. Our findings suggest that in T. cruzi acute infection, when TNF-α is complexed with fibronectin, it favours the altered migration of thymocytes, promoting the release of mature and immature T cells to different compartments of the immune system. Conceptually, this work reinforces the notion that thymocyte migration is a multivectorial biological event in health and disease, and that TNF-α is a further player in the process.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Faculty of Medical Sciences, Institute of Immunology, National University of Rosario, Rosario, Argentina.
| | | | | | | | | | | | | |
Collapse
|
8
|
Pérez AR, Roggero E, Nicora A, Palazzi J, Besedovsky HO, Del Rey A, Bottasso OA. Thymus atrophy during Trypanosoma cruzi infection is caused by an immuno-endocrine imbalance. Brain Behav Immun 2007; 21:890-900. [PMID: 17412557 DOI: 10.1016/j.bbi.2007.02.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Revised: 02/16/2007] [Accepted: 02/20/2007] [Indexed: 02/02/2023] Open
Abstract
C57BL/6 mice infected with Trypanosoma cruzi, the causal agent of Chagas' disease, develop severe thymocyte depletion paralleled by an inflammatory syndrome mediated by tumor necrosis factor-alpha (TNF-alpha). The exacerbated inflammatory reaction induces the activation of hypothalamus-pituitary-adrenal (HPA) axis with the consequent release of corticosterone (CT) into the circulation as a protective response. Thymocyte apoptosis has been related to a rise in TNF-alpha and CT levels, and both mediators are increased in T. cruzi-infected C57BL/6 mice. The depletion of immature CD4(+)CD8(+) thymocytes by apoptosis following infection with the parasite was still present in mice defective in both types of TNF-receptors (double knockout). However, thymic atrophy was prevented by adrenalectomy combined with RU486 administration, demonstrating that this is a CT-driven phenomenon. Our results put emphasis on the importance of an appropriated immuno-endocrine balance during T. cruzi infection and show that functional deviations in the immuno-endocrine equilibrium have profound effects on the thymus and disease outcome.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Instituto de Inmunología, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Santa Fe 3100 (2000) Rosario, Argentina
| | | | | | | | | | | | | |
Collapse
|
9
|
Savino W, Villa-Verde DMS, Mendes-da-Cruz DA, Silva-Monteiro E, Perez AR, Aoki MDP, Bottasso O, Guiñazú N, Silva-Barbosa SD, Gea S. Cytokines and cell adhesion receptors in the regulation of immunity to Trypanosoma cruzi. Cytokine Growth Factor Rev 2007; 18:107-24. [PMID: 17339126 DOI: 10.1016/j.cytogfr.2007.01.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pathophysiology of Chagas' disease is not completely defined, although innate and adaptative immune responses are crucial. In acute infection some parasite antigens can activate macrophages, and this may result in pro-inflammatory cytokine production, nitric oxide synthesis, and consequent control of parasitemia and mortality. Cell-mediated immunity in Trypanosoma cruzi infection is also modulated by cytokines, but in addition to parasite-specific responses, autoimmunity can be also triggered. Importantly, cytokines may also play a role in the cell-mediated immunity of infected subjects. Finally, leukocyte influx towards target tissues is regulated by cytokines, chemokines, and extracellular matrix components which may represent potential therapeutic targets in infected patients. Here we will discuss recent findings on the role of cytokines, chemokines and extracellular matrix components in the regulation of innate and adaptive immunity during T. cruzi infection.
Collapse
Affiliation(s)
- Wilson Savino
- Laboratory of Thymus Research, Department of Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Ave. Brasil 4365, Manguinhos, 21045-900 Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Infectious disease immunology has largely focused on the effector immune response, changes in the blood and peripheral lymphoid organs of infected individuals, and vaccine development. Studies of the thymus in infected individuals have been neglected, although this is progressively changing. The thymus is a primary lymphoid organ, able to generate mature T cells that eventually colonize secondary lymphoid organs, and is therefore essential for peripheral T cell renewal. Recent data show that normal thymocyte development and export can be altered as a result of an infectious disease. One common feature is the severe atrophy of the infected organ, mainly due to the apoptosis-related depletion of immature CD4+CD8+ thymocytes. Additionally, thymocyte proliferation is frequently diminished. The microenvironmental compartment of the thymus is also affected, particularly in acute infectious diseases, with a densification of the epithelial network and an increase in the deposition of extracellular matrix. In the murine model of Chagas disease, intrathymic chemokine production is also enhanced, and thymocytes from Trypanosoma cruzi-infected mice exhibit greater numbers of cell migration-related receptors for chemokines and extracellular matrix, as well as increased migratory responses to the corresponding ligands. This profile is correlated with the appearance of potentially autoreactive thymus-derived immature CD4+CD8+ T cells in peripheral organs of infected animals. A variety of infectious agents—including viruses, protozoa, and fungi—invade the thymus, raising the hypothesis of the generation of central immunological tolerance for at least some of the infectious agent-derived antigens. It seems clear that the thymus is targeted in a variety of infections, and that such targeting may have consequences on the behavior of peripheral T lymphocytes. In this context, thymus-centered immunotherapeutic approaches potentially represent a new tool for the treatment of severe infectious diseases.
Collapse
Affiliation(s)
- Wilson Savino
- Laboratory on Thymus Research, Department of Immunology, Oswaldo Cruz Institute, Inserm-Fiocruz Associated Laboratory of Immunology, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
11
|
Basso B, Cervetta L, Moretti E, Carlier Y, Truyens C. Acute Trypanosoma cruzi infection: IL-12, IL-18, TNF, sTNFR and NO in T. rangeli-vaccinated mice. Vaccine 2004; 22:1868-72. [PMID: 15121297 DOI: 10.1016/j.vaccine.2003.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2003] [Revised: 10/17/2003] [Accepted: 11/12/2003] [Indexed: 01/12/2023]
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.
Collapse
Affiliation(s)
- B Basso
- Facultad de Ciencias Médicas, Universidad Nacional de Córdoba y Servicio Nacional de Chagas, Córdoba, Argentina
| | | | | | | | | |
Collapse
|
12
|
Mjihdi A, Truyens C, Detournay O, Carlier Y. Systemic and placental productions of tumor necrosis factor contribute to induce fetal mortality in mice acutely infected with Trypanosoma cruzi. Exp Parasitol 2004; 107:58-64. [PMID: 15208038 DOI: 10.1016/j.exppara.2004.03.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2003] [Revised: 03/01/2004] [Accepted: 03/30/2004] [Indexed: 11/28/2022]
Abstract
Blood levels and placental productions of IFN-gamma and TNF, known to be harmful for pregnancy, were determined in pregnant mice acutely infected with Trypanosoma cruzi and suffering massive fetal losses without congenital infection. INF-gamma was detected mainly at day 9 and TNF at days 17 and 19 of pregnancy in plasma of infected mice. TNF levels were significantly correlated to the percentages of dead fetuses. Placental cells produced TNF but not IFN-gamma, and addition of T. cruzi lysate to such cells strongly stimulated TNF production. Treatment of infected mice with pentoxifylline, known to decrease IFN-gamma production and to inhibit the TNF-alpha gene transcription, reduced the placental production of TNF, and the fetal mortality in comparison to control animals. Altogether these result suggest that TNF produced at systemic and placental levels plays a role in the fetal mortality induced in mice acutely infected with T. cruzi.
Collapse
Affiliation(s)
- Abdelkarim Mjihdi
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | |
Collapse
|
13
|
Cardoni RL, García MM, De Rissio AM. Proinflammatory and anti-inflammatory cytokines in pregnant women chronically infected with Trypanosoma cruzi. Acta Trop 2004; 90:65-72. [PMID: 14739025 DOI: 10.1016/j.actatropica.2003.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mother-to-child transmission of intracellular parasites could be related to the production of immunoregulatory cytokines. The levels of gamma interferon (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha) and lnterleukin (IL)-10 were evaluated during pregnancy in sera of women chronically infected with Trypanosoma cruzi that delivered infected or non-infected children. The levels of IL-10 increased in both, women only pregnant and only infected, compared to non-infected non-pregnant women. However, in pregnant women chronically infected with T. cruzi, IL-10 did not increase significantly, neither in the mothers of infected nor in the mothers of non-infected children. The levels of the inflammatory cytokine TNF-alpha were not affected in normal pregnancy but increased in the infected mothers of non-infected children. The levels of IFN-gamma did not increase in the groups studied, indicating that the production of this pro-inflammatory cytokine was controlled, even when the levels of IL-10 did not increase, as in pregnant women chronically infected with T. cruzi.
Collapse
Affiliation(s)
- R L Cardoni
- Instituto Nacional de Parasitdogía (INP) "Dr M. Fatala Chabén", ANLIS "C.G. Malbrán", Av. Paseo Colón 568, 1063, Buenos Aires, Argentina.
| | | | | |
Collapse
|
14
|
Mjihdi A, Lambot MA, Stewart IJ, Detournay O, Noël JC, Carlier Y, Truyens C. Acute Trypanosoma cruzi infection in mouse induces infertility or placental parasite invasion and ischemic necrosis associated with massive fetal loss. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:673-80. [PMID: 12163392 PMCID: PMC1850751 DOI: 10.1016/s0002-9440(10)64223-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
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 in Latin America, on reproduction of mice. Although mating of infected mice occurred at a normal rate, 80% of them did not become gravid. In the few gravid infected mice, implantation numbers were as in uninfected control mice, but 28% of fetuses resorbed. Such infertility and early fetal losses were significantly associated with high maternal parasitemia. The remaining fetuses presented with reduced weights and all died later in gestation or within 48 hours after birth. Several organs of these fetuses were infiltrated by polynuclear cells and presented ischemic necrosis but did not harbor T. cruzi parasites, discarding congenital infection as the cause of mortality. However, surprisingly, the deciduas were massively invaded by T. cruzi parasites, harboring 125-fold more amastigotes than the maternal heart or other placental tissues. Parasites were significantly more numerous in the placentas of dead fetuses. In addition, placentas contained inflammatory infiltrates and displayed ischemic necrosis, fibrin deposits, and vascular thromboses. These results show that acute T. cruzi infection totally impairs reproduction in mice through inducing infertility or fetal-neonatal losses in association with placental parasite invasion and ischemic necrosis.
Collapse
Affiliation(s)
- Abdelkarim Mjihdi
- Laboratoire de Parasitologie, Faculté de Médecine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
15
|
Vekemans J, Truyens C, Torrico F, Solano M, Torrico MC, Rodriguez P, Alonso-Vega C, Carlier Y. Maternal Trypanosoma cruzi infection upregulates capacity of uninfected neonate cells To produce pro- and anti-inflammatory cytokines. Infect Immun 2000; 68:5430-4. [PMID: 10948177 PMCID: PMC101811 DOI: 10.1128/iai.68.9.5430-5434.2000] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
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.
Collapse
Affiliation(s)
- J Vekemans
- Laboratory of Parasitology, Faculty of Medicine, University of Brussels, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Fournet A, Rojas de Arias A, Ferreira ME, Nakayama H, Torres de Ortiz S, Schinini A, Samudio M, Vera de Bilbao N, Lavault M, Bonté F. Efficacy of the bisbenzylisoquinoline alkaloids in acute and chronic Trypanosoma cruzi murine model. Int J Antimicrob Agents 2000; 13:189-95. [PMID: 10724023 DOI: 10.1016/s0924-8579(99)00117-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have shown previously that daphnoline and cepharanthine are active against Trypanosoma cruzi and inhibited trypanothione reductase. The effects of oral treatments with daphnoline, cepharanthine and benznidazole were examined in Balb/c mice infected with T. cruzi acutely and chronically. In acute infections, parasitaemia was significantly reduced in the daphnoline-treated mice compared with controls and benznidazole-treated mice. The parasitological cure rate was increased in mice treated with daphnoline. Fifty days after infection, the negative serological response in both models was significantly different for the three tested drugs. Daphnoline showed the highest negative serological rate (48%). In chronically infected mice treated with daphnoline, we were unable to detect parasites in 70% of mice. The results obtained of oral treatment of daphnoline suggest that this bisbenzylisoquinoline may be useful in the treatment of acute and chronic Chagas' disease. This was not seen with cepharanthine, an excellent trypanothione reductase inhibitor.
Collapse
Affiliation(s)
- A Fournet
- Institut de Recherche pour le Développement, ex-ORSTOM, UR Sante, Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chandrasekar B, Melby PC, Troyer DA, Colston JT, Freeman GL. Temporal expression of pro-inflammatory cytokines and inducible nitric oxide synthase in experimental acute Chagasic cardiomyopathy. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 152:925-34. [PMID: 9546353 PMCID: PMC1858258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To characterize the kinetics of myocardial cytokine and inducible nitric oxide synthase (iNOS) expression in acute Chagasic cardiomyopathy, we studied a rat model of acute Trypanosoma cruzi infection. Rats were euthanized 36 hours and 5, 10, and 15 days after infection, and hearts were collected for histology, mRNA, and protein analyses. Histological analysis of myocardium showed a progressive increase in the number of amastigotes and mononuclear inflammatory cells. Organisms were first detected 5 days after intraperitoneal inoculation as isolated nests and became numerous by day 15. Northern blot analysis of total RNA revealed no signal for interleukin (IL)-1beta or tumor necrosis factor (TNF)-alpha and a weak signal for IL-6 in control hearts. High levels of expression for the three genes were detected in the infected animals at 36 hours after infection. Although IL-1beta and IL-6 levels increased steadily up to 10 days, TNF-alpha levels were the highest at 5 days, remained high at 10 days, and declined thereafter. Western blot analysis showed similar results to that of mRNA expression. No signal was detected for iNOS in the controls, but both its mRNA and protein were found in the infected animals, with levels being highest at 15 days after infection. Immunohistochemistry revealed no iNOS immunoreactivity in uninfected animals, but intense iNOS staining was detected in blood vessels of infected animals, which decreased progressively with period of infection. Positive staining for iNOS in cardiomyocytes was first detected at 36 hours after infection (at a time when there was no histological inflammatory reaction), which steadily increased, being the highest at 15 days after infection. These results indicate that, in addition to mechanical damage by T. cruzi, substantial pro-inflammatory cytokine production within the myocardium is likely to participate in the pathophysiology of acute Chagasic cardiomyopathy.
Collapse
Affiliation(s)
- B Chandrasekar
- Department of Medicine, The University of Texas Health Science Center, South Texas Veterans Health Care System, San Antonio 78284-7872, USA
| | | | | | | | | |
Collapse
|
18
|
Feldman S, Revelli S, Davila H, Marcipar A, Rojas M, Avila JL, Bottasso OA. Depressed autoantibody synthesis in Trypanosoma cruzi-infected rats born to mothers undergoing this infection during pregnancy. J Reprod Immunol 1997; 34:177-84. [PMID: 9350635 DOI: 10.1016/s0165-0378(97)00045-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Earlier work indicated that Trypanosoma cruzi infection in pregnant rats decreased the amount of myocardial damage that developed in their chronically infected offspring. Given the suspected role of autoimmune mechanisms in the generation of chronic myocarditis, we evaluated whether this maternal intervention was likely to affect the synthesis of autoantibodies in infected young. Autoantibodies were investigated against molecules exhibiting cross-reactivity with T. cruzi antigens or not, that is cerebroside sulphate (sulphatide) and actin, respectively. Female '1' rats (75 days old) that had been mated with syngeneic sires were separated into two groups, one challenged with living trypomastigotes at 7, 14 and 21 days following mating, and the other one given physiologic saline at the same intervals. At the time of weaning, offspring were injected with 10(6)/T. cruzi to constitute two infected groups: young born to infected mothers (InMoTc) and young delivered by uninfected mothers (CoMoTc). Serum antibodies were investigated by ELISA at 30 and 60 days post-infection, which represents acute and chronic infection, respectively. T. cruzi infection was associated with the production of anti-sulphatide antibodies, but the phenomenon was significantly less evident in InMoTc young and virtually unnoticeable during their chronic infection. Unlike the anti-sulphatide results, levels of anti-actin antibodies showed no differences between CoMoTc and InMoTc rats when compared during acute or chronic infection. The decreased production of anti-sulphatide autoantibodies of InMoTc offspring may be due to a modification of the immune repertoire of offspring because of the contact with parasite antigens during ontogeny.
Collapse
Affiliation(s)
- S Feldman
- Cátedra de Química Biologica, Facultad de Ciencias, Médicas de Rosario, Argentina
| | | | | | | | | | | | | |
Collapse
|
19
|
Tarleton RL, Zhang L, Downs MO. "Autoimmune rejection" of neonatal heart transplants in experimental Chagas disease is a parasite-specific response to infected host tissue. Proc Natl Acad Sci U S A 1997; 94:3932-7. [PMID: 9108082 PMCID: PMC20545 DOI: 10.1073/pnas.94.8.3932] [Citation(s) in RCA: 106] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/1996] [Accepted: 02/05/1997] [Indexed: 02/04/2023] Open
Abstract
Infection with the protozoan parasite Trypanosoma cruzi often results in chronic heart- and gut-associated disease known as Chagas disease. In this study we show that contrary to previous reports, neonatal hearts transplanted into mice chronically infected with T. cruzi do not exhibit signs of autoimmune-type rejection or any significant inflammatory response. In addition to an absence of inflammation, these syngeneic heart transplants survive for more than 1 year and are absolutely free of parasites as determined by in situ PCR analysis. However, if well-established transplanted hearts in chronically infected mice are directly injected with live parasites, a rapid and dramatic inflammatory response ensues that results in cessation of heart function. Likewise, transplanted hearts established in mice prior to systemic infection with T. cruzi or hearts transplanted into mice during the acute stage of T. cruzi infection become parasitized and develop inflammatory foci. In these cases where the transplanted hearts become parasitized, the ensuing inflammatory response is nearly identical to that observed in the native hearts of T. cruzi-infected mice in terms of cell types present and adhesion molecules and cytokines expressed. Importantly, this response is strikingly different from that observed in the allogeneic heart rejection. These results clearly document that parasitization of heart tissue is both necessary and sufficient for the induction of tissue damage in Chagas disease and strongly argue against a principal autoimmune etiology for this disease.
Collapse
Affiliation(s)
- R L Tarleton
- Department of Cellular Biology, The University of Georgia, Athens 30602, USA
| | | | | |
Collapse
|
20
|
Fournet A, Ferreira ME, de Arias AR, Schinini A, Nakayama H, Torres S, Sanabria R, Guinaudeau H, Bruneton J. The effect of bisbenzylisoquinoline alkaloids on Trypanosoma cruzi infections in mice. Int J Antimicrob Agents 1997; 8:163-70. [DOI: 10.1016/s0924-8579(97)00373-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/1996] [Indexed: 11/25/2022]
|