1
|
da Silva RJ, Almeida MPO, Gomes AO, Franco PS, de Souza G, Rosini AM, Milian ICB, Servato JPS, Mineo JR, Mineo TWP, Silva NM, Ferro EAV, Barbosa BF. Intercellular adhesion molecule (ICAM)-1 is required to control Toxoplasma gondii infection in uterine tissues and establish a successful gestation in a murine model of congenital toxoplasmosis. Placenta 2025:S0143-4004(25)00103-1. [PMID: 40204594 DOI: 10.1016/j.placenta.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
The placenta acts as a critical barrier against pathogens during pregnancy, although Toxoplasma gondii can breach this defense, leading to congenital infections. Intercellular adhesion molecule-1 (ICAM-1) is an adhesion molecule involved in immune responses, including leukocyte recruitment and pathogen clearance. Here, we investigate the role of ICAM-1 in gestational success and T. gondii infection using wild-type (WT) and ICAM-1 knockout (ICAM-1-/-) mice across early, mid- and late pregnancy stages. In early pregnancy, ICAM-1-/- mice infected with T. gondii exhibited a significantly higher embryonic loss rate (63 %) compared to WT mice (5 %). This was accompanied by an increased parasite burden in uterine tissues and elevated systemic and local IFN-γ levels, despite a reduced local inflammatory response. In contrast, mid-pregnancy showed no significant differences in fetal loss or implantation success among groups, suggesting ICAM-1 plays a limited role at this stage. During late pregnancy, ICAM-1-/- mice experienced higher embryonic loss rates (40 %) compared to WT mice (26.2 %), along with reduced implantation success and elevated IFN-γ levels, though parasite burden remained unchanged. Histological analysis revealed a less severe inflammatory profile in infected ICAM-1-/- uterine tissues, marked by reduced necrosis and hyperemia compared to WT mice. FOXP3 expression, a marker of regulatory T cells, was unaffected by ICAM-1, although a trend towards reestablishment was observed in infected ICAM-1-/- mice. Our findings underscore the critical role of ICAM-1 in ensuring gestational success during T. gondii infection, particularly in early pregnancy, by modulating immune responses at the maternal-fetal interface.
Collapse
Affiliation(s)
- Rafaela José da Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Marcos Paulo Oliveira Almeida
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Angelica Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil.
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Iliana Claudia Balga Milian
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | | | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
2
|
Teixeira SC, Melo Fernandes TAD, Souza GD, Rosini AM, Fajardo Martínez AF, Gomes AO, Alves RN, Lopes DS, Silva MVD, Beraldo-Neto E, Clissa PB, Barbosa BF, Ávila VDMR, Ferro EAV. MjTX-II, a Lys49-PLA 2 from Bothrops moojeni snake venom, restricts Toxoplasma gondii infection via ROS and VEGF regulation. Chem Biol Interact 2025; 409:111417. [PMID: 39922520 DOI: 10.1016/j.cbi.2025.111417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Owing to the lack of efficient therapy and emerging resistance strains, toxoplasmosis affects about one-third of the world's population. Also, pregnancy-related infection can cause vertical transmission and result in fetal death. Despite the global efforts to combat Toxoplasma gondii infection, conventional therapies have been associated with serious side effects. Therefore, it is relevant to search for effective and less-toxic treatments of toxoplasmosis. In this scenario, snake venoms emerged as a promising source of therapeutic molecules due to their wide variety of biological effects. The present study investigated the anti-T. gondii effects of MjTX-II, a Lys49-PLA2 isolated from Bothrops moojeni, in trophoblast cells and villous explants from the third trimester of pregnancy. We found that non-cytotoxic doses of MjTX-II impaired parasite invasion and intracellular growth in BeWo cells. Also, MjTX-II-pre-treated T. gondii tachyzoites exhibited irregular rough surfaces, papules, and dimples, suggesting a possible action directly on the parasites. Moreover, MjTX-II was able to modulate the host environment by increasing ROS and cytokine levels involved in the control of infection. In addition, we observed that MjTX-II decreased VEGF levels and the addition of rVEGF increased T. gondii growth in BeWo cells. Through molecular docking simulations, we verified that MjTX-II is able to bind VEGFR2 and ICAM-1 receptors associated with parasite proliferation and dissemination. This work contributes to the discovery of therapeutic targets against T. gondii infection and for the development of effective and low-toxic antiparasitic molecules against congenital toxoplasmosis.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | - Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Aryani Felixa Fajardo Martínez
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Angelica Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Rosiane Nascimento Alves
- Department of Agricultural and Natural Science, Universidade do Estado de Minas Gerais, Ituiutaba, MG, Brazil
| | - Daiana Silva Lopes
- Institute Multidisciplinary in Health, Universidade Federal da Bahia, Vitória da Conquista, BA, Brazil
| | | | | | | | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Laboratory of Biochemistry and Animal Toxins, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
3
|
Castillo C, Díaz-Luján C, Liempi A, Fretes R, Kemmerling U. Mammalian placental explants: A tool for studying host-parasite interactions and placental biology. Placenta 2024:S0143-4004(24)00291-1. [PMID: 38910051 DOI: 10.1016/j.placenta.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
The placenta plays a critical role in host-pathogen interactions. Thus, ex vivo infection of mammalian placental explants is an excellent and simple method to study the mechanisms of cellular and tissue invasion by different pathogens in different mammalian species. These explants can be maintained in culture for several days, preserving the tissue architecture and resembling in-utero conditions under more physiological conditions than their isolated counterparts in isolated cell culture models. In addition, placental explants not only allow us to study how the placenta responds and defends itself against various infections but also provide a versatile platform for advancing our understanding of placental biology and the immune response. Furthermore, they serve as powerful tools for drug discovery, facilitating the screening of potential therapeutics for placental infections and for the identification of diagnostic markers. This review highlights the utility of mammalian placental explants in studying the host-pathogen interaction of two relevant protozoan parasites, Trypanosoma cruzi, the causative agent of Chagas disease, and Toxoplasma gondii, the etiological agent of Toxoplasmosis. Here, we discuss the different methodologies and technical aspects of the model, as well as the effect of both parasites on placental responses in human, canine, and ovine explants.
Collapse
Affiliation(s)
- Christian Castillo
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Cintia Díaz-Luján
- Institute and Cathedra of Cell Biology, Histology and Embryology, Health Science Faculty, INICSA (CONICET)-Universidad Nacional de Córdoba and Villa María, Córdoba, Argentina
| | - Ana Liempi
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ricardo Fretes
- Institute and Cathedra of Cell Biology, Histology and Embryology, Health Science Faculty, INICSA (CONICET)-Universidad Nacional de Córdoba and Villa María, Córdoba, Argentina
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
4
|
Souza Silva VR, Mota CM, Maia LP, Ferreira FB, Miranda VDS, Silva NM, Ferro EAV, Mineo JR, Mineo TWP. Macrophage migration inhibitory factor favors Neospora caninum infection in mice. Microb Pathog 2024; 189:106577. [PMID: 38367848 DOI: 10.1016/j.micpath.2024.106577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/19/2024]
Abstract
Neospora caninum is a protozoan parasite with worldwide incidence, acting as a major cause of reproductive failures in ruminants and neuromuscular symptoms in dogs. Macrophage Migration Inhibitory Factor (MIF) is produced by several cell types and exhibits a central role in immune responses against intracellular pathogens. The present study aimed to comprehend the role of MIF in the relationship between N. caninum and its host. We used in vivo, in vitro and ex vivo experiments in a model of infection based on genetically deficient mice to analyze the infection kinetics and inflammatory markers. MIF production was measured in response to N. caninum during the acute and chronic phases of the infection. While Mif-/- mice survived lethal doses of NcLiv tachyzoites, sublethal infections in these mice showed that parasite burden was controlled in target tissues, alongside with reduced inflammatory infiltrates detected in lung and brain sections. TNF was increased at the initial site of the infection in genetically deficient mice and the MIF-dependent reduction was confirmed in vitro with macrophages and ex vivo with primed spleen cells. In sum, MIF negatively regulated host immunity against N. caninum, favoring disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eloísa A Vieira Ferro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia. Av. Amazonas, Campus Umuarama., 38405-320, Uberlândia, Minas Gerais, Brazil
| | | | | |
Collapse
|
5
|
Teixeira SC, Paschoalino M, de Souza G, Rosini AM, de Lima Junior JP, Luz LC, Fajardo Martínez AF, Alves RN, Almeida MPO, Damasceno JL, Silva MJB, Ietta F, Barbosa BF, Ferro EAV, Gomes Martins CH. Rottlerin impairs early and late steps of Toxoplasma gondii infection in human trophoblast cells and villous explants. Chem Biol Interact 2023; 384:110716. [PMID: 37722575 DOI: 10.1016/j.cbi.2023.110716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Congenital toxoplasmosis, caused by the opportunistic protozoan parasite T. gondii, can cause stillbirths, miscarriages and fetal abnormalities, as well as encephalitis and chorioretinitis in newborns. Available treatment options rely on antiparasitic drugs that have been linked to serious side effects, high toxicity and the development of drug-resistant parasites. The search for alternative therapeutics to treat this disease without acute toxicity for the mother and child is essential for the advancement of current therapeutic procedures. The present study aimed to unravel the mode of the anti-T. gondii action of Rottlerin, a natural polyphenol with multiple pharmacological properties described. Herein, we further assessed the antiparasitic activity of Rottlerin against T. gondii infection on the human trophoblastic cells (BeWo cells) and, for the first time, on human villous explants. We found that non-cytotoxic doses of Rottlerin impaired early and late steps of parasite infection with an irreversible manner in BeWo cells. Rottlerin caused parasite cell cycle arrest in G1 phase and compromised the ability of tachyzoites to infect new cells, thus highlighting the possible direct action on parasites. An additional and non-exclusive mechanism of action of Rottlerin involves the modulation of host cell components, by affecting lipid droplet formation, mitochondrial function and upregulation of the IL-6 and MIF levels in BeWo cells. Supporting our findings, Rottlerin also controlled T. gondii proliferation in villous explants with low toxicity and reduced the IL-10 levels, a cytokine associated with parasite susceptibility. Collectively, our results highlighted the potential use of Rottlerin as a promising tool to prevent and/or treat congenital toxoplasmosis.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marina Paschoalino
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Joed Pires de Lima Junior
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Luana Carvalho Luz
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Aryani Felixa Fajardo Martínez
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Rosiane Nascimento Alves
- Department of Agricultural and Natural Sciences, Universidade do Estado de Minas Gerais, Ituiutaba, MG, Brazil
| | - Marcos Paulo Oliveira Almeida
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Jaqueline Lopes Damasceno
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marcelo José Barbosa Silva
- Department of Immunology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| | - Carlos Henrique Gomes Martins
- Laboratory of Antimicrobial Testing, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
6
|
Teixeira SC, Rosini AM, de Souza G, Fajardo Martínez AF, Silva RJ, Ambrósio SR, Sola Veneziani RC, Bastos JK, Gomes Martins CH, Barbosa BF, Vieira Ferro EA. Polyalthic acid and oleoresin from Copaifera trapezifolia Hayne reduce Toxoplasma gondii growth in human villous explants, even triggering an anti-inflammatory profile. Exp Parasitol 2023; 250:108534. [PMID: 37100271 DOI: 10.1016/j.exppara.2023.108534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023]
Abstract
Due to the lack of efficient antiparasitic therapy and vaccines, as well as emerging resistance strains, congenital toxoplasmosis is still a public health issue worldwide. The present study aimed to assess the effects of an oleoresin obtained from the species Copaifera trapezifolia Hayne (CTO), and an isolated molecule found in the CTO, ent-polyalthic acid (ent-15,16-epoxy-8(17),13(16),14-labdatrien-19-oic acid) (named as PA), against T. gondii infection. We used human villous explants as an experimental model of human maternal-fetal interface. Uninfected and infected villous explants were exposed to the treatments; the parasite intracellular proliferation and the cytokine levels were measured. Also, T. gondii tachyzoites were pre-treated and the parasite proliferation was determined. Our findings showed that CTO and PA reduced efficiently the parasite growth with an irreversible action, but without causing toxicity to the villi. Also, treatments reduced the levels of IL-6, IL-8, MIF and TNF by villi, what configures a valuable treatment option for the maintenance of a pregnancy in an infectious context. In addition to a possible direct effect on parasites, our data suggest an alternative mechanism by which CTO and PA alter the villous explants environment and then impair parasite growth, since the pre-treatment of villi resulted in lower parasitic infection. Here, we highlighted PA as an interesting tool for the design of new anti-T. gondii compounds.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Aryani Felixa Fajardo Martínez
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Sergio Ricardo Ambrósio
- Nucleus of Research in Technological and Exact Sciences, University of Franca, Franca, SP, Brazil.
| | | | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Carlos Henrique Gomes Martins
- Department of Microbiology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
7
|
Faral-Tello P, Pagotto R, Bollati-Fogolín M, Francia ME. Modeling the human placental barrier to understand Toxoplasma gondii´s vertical transmission. Front Cell Infect Microbiol 2023; 13:1130901. [PMID: 36968102 PMCID: PMC10034043 DOI: 10.3389/fcimb.2023.1130901] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
Toxoplasma gondii is a ubiquitous apicomplexan parasite that can infect virtually any warm-blooded animal. Acquired infection during pregnancy and the placental breach, is at the core of the most devastating consequences of toxoplasmosis. T. gondii can severely impact the pregnancy’s outcome causing miscarriages, stillbirths, premature births, babies with hydrocephalus, microcephaly or intellectual disability, and other later onset neurological, ophthalmological or auditory diseases. To tackle T. gondii’s vertical transmission, it is important to understand the mechanisms underlying host-parasite interactions at the maternal-fetal interface. Nonetheless, the complexity of the human placenta and the ethical concerns associated with its study, have narrowed the modeling of parasite vertical transmission to animal models, encompassing several unavoidable experimental limitations. Some of these difficulties have been overcome by the development of different human cell lines and a variety of primary cultures obtained from human placentas. These cellular models, though extremely valuable, have limited ability to recreate what happens in vivo. During the last decades, the development of new biomaterials and the increase in stem cell knowledge have led to the generation of more physiologically relevant in vitro models. These cell cultures incorporate new dimensions and cellular diversity, emerging as promising tools for unraveling the poorly understood T. gondii´s infection mechanisms during pregnancy. Herein, we review the state of the art of 2D and 3D cultures to approach the biology of T. gondii pertaining to vertical transmission, highlighting the challenges and experimental opportunities of these up-and-coming experimental platforms.
Collapse
Affiliation(s)
- Paula Faral-Tello
- Laboratory of Apicomplexan Biology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Romina Pagotto
- Cell Biology Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Maria E. Francia
- Laboratory of Apicomplexan Biology, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Parasitología y Micología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- *Correspondence: Maria E. Francia,
| |
Collapse
|
8
|
Guimarães Gois PS, Franco PS, Cota Teixeira S, Guirelli PM, de Araújo TE, da Fonseca Batistão DW, de Oliveira FC, Lícia Santos Ferreira G, de Oliveira Gomes A, Favoreto S, Mineo JR, de Freitas Barbosa B, Ferro EAV. Polarisation of human macrophages towards an M1 subtype triggered by an atypical Brazilian strain of Toxoplasma gondii results in a reduction in parasite burden. Folia Parasitol (Praha) 2022; 69. [DOI: 10.14411/fp.2022.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 06/16/2022] [Indexed: 11/19/2022]
|
9
|
Rosini AM, Teixeira SC, Milian ICB, Silva RJ, de Souza G, Luz LC, Gomes AO, Mineo JR, Mineo TWP, Ferro EAV, Barbosa BF. LPS-mediated activation of TLR4 controls Toxoplasma gondii growth in human trophoblast cell (BeWo) and human villous explants in a dependent-manner of TRIF, MyD88, NF-κB and cytokines. Tissue Cell 2022; 78:101907. [DOI: 10.1016/j.tice.2022.101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/22/2022] [Accepted: 08/22/2022] [Indexed: 02/07/2023]
|
10
|
Transforming growth factor (TGF)-β1 and interferon (IFN)-γ differentially regulate ICAM-1 expression and adhesion of Toxoplasma gondii to human trophoblast (BeWo) and uterine cervical (HeLa) cells. Acta Trop 2021; 224:106111. [PMID: 34450063 DOI: 10.1016/j.actatropica.2021.106111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 07/24/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022]
Abstract
Toxoplasma gondii is a parasite able to infect various cell types, including trophoblast cells. Studies have demonstrated that interleukin (IL)-10, transforming growth factor (TGF)-β1 and interferon (IFN)-γ are involved in the susceptibility of BeWo trophoblast cells to T. gondii infection. Furthermore, T. gondii is able to adhere to the plasma membrane of host cells through intercellular adhesion molecule (ICAM)-1. Thus, the present study aimed to assess the role of IL-10, TGF-β1 and IFN-γ in the expression of ICAM-1 in BeWo and HeLa cells and to analyze the role of ICAM-1 in the adhesion and invasion of T. gondii to these cells under the influence of these cytokines. For this purpose, BeWo and HeLa cells were treated or not, before and after T. gondii infection, with rIL-10, rTGF-β1 or rIFN-γ. For the BeWo cells, rIL-10 and rTGF-β1 favored susceptibility to infection, but only rTGF-β1 and rIFN-γ increased ICAM-1 expression, and TNF-α release. On the other hand, rIFN-γ downregulated the expression of ICAM-1 triggered by T. gondii in HeLa cells, leading to control of the infection. Moreover, we observed that upregulation of ICAM-1, mediated by cytokine's stimulation, in BeWo and HeLa cells resulted in a high number rate of both parasite adhesion and invasion to these cells, which were strongly reduced after ICAM-1 neutralization. Likewise, the blockage of ICAM-1 molecule also impaired T. gondii infection in human villous explants. Taken together, these findings demonstrate that TGF-β1 and IFN-γ differentially regulate ICAM-1 expression, which may interfere in the adhesion/invasion of T. gondii to BeWo and HeLa cells for modulating susceptibility to infection.
Collapse
|
11
|
Rojas-Pirela M, Medina L, Rojas MV, Liempi AI, Castillo C, Pérez-Pérez E, Guerrero-Muñoz J, Araneda S, Kemmerling U. Congenital Transmission of Apicomplexan Parasites: A Review. Front Microbiol 2021; 12:751648. [PMID: 34659187 PMCID: PMC8519608 DOI: 10.3389/fmicb.2021.751648] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Apicomplexans are a group of pathogenic protists that cause various diseases in humans and animals that cause economic losses worldwide. These unicellular eukaryotes are characterized by having a complex life cycle and the ability to evade the immune system of their host organism. Infections caused by some of these parasites affect millions of pregnant women worldwide, leading to various adverse maternal and fetal/placental effects. Unfortunately, the exact pathogenesis of congenital apicomplexan diseases is far from being understood, including the mechanisms of how they cross the placental barrier. In this review, we highlight important aspects of the diseases caused by species of Plasmodium, Babesia, Toxoplasma, and Neospora, their infection during pregnancy, emphasizing the possible role played by the placenta in the host-pathogen interaction.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.,Facultad de Farmacia y Bioanálisis, Universidad de Los Andes, Mérida, Venezuela
| | - Lisvaneth Medina
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Maria Verónica Rojas
- Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Ana Isabel Liempi
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Christian Castillo
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago, Chile
| | | | - Jesús Guerrero-Muñoz
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sebastian Araneda
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Facultad de Salud y Odontología, Universidad Diego Portales, Santiago, Chile
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
12
|
de Souza G, Silva RJ, Milián ICB, Rosini AM, de Araújo TE, Teixeira SC, Oliveira MC, Franco PS, da Silva CV, Mineo JR, Silva NM, Ferro EAV, Barbosa BF. Cyclooxygenase (COX)-2 modulates Toxoplasma gondii infection, immune response and lipid droplets formation in human trophoblast cells and villous explants. Sci Rep 2021; 11:12709. [PMID: 34135407 PMCID: PMC8209052 DOI: 10.1038/s41598-021-92120-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
Congenital toxoplasmosis is represented by the transplacental passage of Toxoplasma gondii from the mother to the fetus. Our studies demonstrated that T. gondii developed mechanisms to evade of the host immune response, such as cyclooxygenase (COX)-2 and prostaglandin E2 (PGE2) induction, and these mediators can be produced/stored in lipid droplets (LDs). The aim of this study was to evaluate the role of COX-2 and LDs during T. gondii infection in human trophoblast cells and villous explants. Our data demonstrated that COX-2 inhibitors decreased T. gondii replication in trophoblast cells and villous. In BeWo cells, the COX-2 inhibitors induced an increase of pro-inflammatory cytokines (IL-6 and MIF), and a decrease in anti-inflammatory cytokines (IL-4 and IL-10). In HTR-8/SVneo cells, the COX-2 inhibitors induced an increase of IL-6 and nitrite and decreased IL-4 and TGF-β1. In villous explants, the COX-2 inhibitors increased MIF and decreased TNF-α and IL-10. Furthermore, T. gondii induced an increase in LDs in BeWo and HTR-8/SVneo, but COX-2 inhibitors reduced LDs in both cells type. We highlighted that COX-2 is a key factor to T. gondii proliferation in human trophoblast cells, since its inhibition induced a pro-inflammatory response capable of controlling parasitism and leading to a decrease in the availability of LDs, which are essentials for parasite growth.
Collapse
Affiliation(s)
- Guilherme de Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Iliana Claudia Balga Milián
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Thádia Evelyn de Araújo
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Mário Cézar Oliveira
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Claudio Vieira da Silva
- Laboratory of Trypanosomatids, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Campus Umuarama, Av. Pará, 1720, Uberlândia, MG, 38405-320, Brazil.
| |
Collapse
|
13
|
Arranz-Solís D, Mukhopadhyay D, Saeij JJP. Toxoplasma Effectors that Affect Pregnancy Outcome. Trends Parasitol 2021; 37:283-295. [PMID: 33234405 PMCID: PMC7954850 DOI: 10.1016/j.pt.2020.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/18/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
As an immune-privileged organ, the placenta can tolerate the introduction of antigens without inducing a strong inflammatory response that would lead to abortion. However, for the control of intracellular pathogens, a strong Th1 response characterized by the production of interferon-γ is needed. Thus, invasion of the placenta by intracellular parasites puts the maternal immune system in a quandary: The proinflammatory response needed to eliminate the pathogen can also lead to abortion. Toxoplasma is a highly successful parasite that causes lifelong chronic infections and is a major cause of abortions in humans and livestock. Here, we discuss how Toxoplasma strain type and parasite effectors influence host cell signaling pathways, and we speculate about how this might affect the outcome of gestation.
Collapse
Affiliation(s)
- David Arranz-Solís
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Debanjan Mukhopadhyay
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Jeroen J P Saeij
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
14
|
Todros T, Paulesu L, Cardaropoli S, Rolfo A, Masturzo B, Ermini L, Romagnoli R, Ietta F. Role of the Macrophage Migration Inhibitory Factor in the Pathophysiology of Pre-Eclampsia. Int J Mol Sci 2021; 22:1823. [PMID: 33673075 PMCID: PMC7917653 DOI: 10.3390/ijms22041823] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
Proinflammatory cytokines are produced in pregnancy in response to the invading pathogens and/or nonmicrobial causes such as damage-associated molecules and embryonic semi-allogenic antigens. While inflammation is essential for a successful pregnancy, an excessive inflammatory response is implicated in several pathologies including pre-eclampsia (PE). This review focuses on the proinflammatory cytokine macrophage migration inhibitory factor (MIF), a critical regulator of the innate immune response and a major player of processes allowing normal placental development. PE is a severe pregnancy-related syndrome characterized by exaggerated inflammatory response and generalized endothelial damage. In some cases, usually of early onset, it originates from a maldevelopment of the placenta, and is associated with intrauterine growth restriction (IUGR) (placental PE). In other cases, usually of late onset, pre-pregnancy maternal diseases represent risk factors for the development of the disease (maternal PE). Available data suggest that low MIF production in early pregnancy could contribute to the abnormal placentation. The resulting placental hypoxia in later pregnancy could produce high release of MIF in maternal serum typical of placental PE. More studies are needed to understand the role of MIF, if any, in maternal PE.
Collapse
Affiliation(s)
- Tullia Todros
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126 Turin, Italy; (T.T.); (A.R.)
| | - Luana Paulesu
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| | - Simona Cardaropoli
- Department of Public Health and Pediatrics, University of Turin, 10126 Turin, Italy;
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126 Turin, Italy; (T.T.); (A.R.)
| | | | - Leonardo Ermini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| | - Roberta Romagnoli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; (L.E.); (R.R.); (F.I.)
| |
Collapse
|
15
|
Pastor-Fernández I, Collantes-Fernández E, Jiménez-Pelayo L, Ortega-Mora LM, Horcajo P. Modeling the Ruminant Placenta-Pathogen Interactions in Apicomplexan Parasites: Current and Future Perspectives. Front Vet Sci 2021; 7:634458. [PMID: 33553293 PMCID: PMC7859336 DOI: 10.3389/fvets.2020.634458] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
Neospora caninum and Toxoplasma gondii are one of the main concerns of the livestock sector as they cause important economic losses in ruminants due to the reproductive failure. It is well-known that the interaction of these parasites with the placenta determines the course of infection, leading to fetal death or parasite transmission to the offspring. However, to advance the development of effective vaccines and treatments, there are still important gaps on knowledge on the placental host-parasite interactions that need to be addressed. Ruminant animal models are still an indispensable tool for providing a global view of the pathogenesis, lesions, and immune responses, but their utilization embraces important economic and ethics restrictions. Alternative in vitro systems based on caruncular and trophoblast cells, the key cellular components of placentomes, have emerged in the last years, but their use can only offer a partial view of the processes triggered after infection as they cannot mimic the complex placental architecture and neglect the activity of resident immune cells. These drawbacks could be solved using placental explants, broadly employed in human medicine, and able to preserve its cellular architecture and function. Despite the availability of such materials is constrained by their short shelf-life, the development of adequate cryopreservation protocols could expand their use for research purposes. Herein, we review and discuss existing (and potential) in vivo, in vitro, and ex vivo ruminant placental models that have proven useful to unravel the pathogenic mechanisms and the host immune responses responsible for fetal death (or protection) caused by neosporosis and toxoplasmosis.
Collapse
Affiliation(s)
| | | | | | | | - Pilar Horcajo
- Animal Health and Zoonoses (SALUVET) Group, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
16
|
Costa IN, Ribeiro M, Silva Franco P, da Silva RJ, de Araújo TE, Milián ICB, Luz LC, Guirelli PM, Nakazato G, Mineo JR, Mineo TWP, Barbosa BF, Ferro EAV. Biogenic Silver Nanoparticles Can Control Toxoplasma gondii Infection in Both Human Trophoblast Cells and Villous Explants. Front Microbiol 2021; 11:623947. [PMID: 33552033 PMCID: PMC7858645 DOI: 10.3389/fmicb.2020.623947] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/31/2020] [Indexed: 01/08/2023] Open
Abstract
The combination of sulfadiazine and pyrimethamine plus folinic acid is the conventional treatment for congenital toxoplasmosis. However, this classical treatment presents teratogenic effects and bone marrow suppression. In this sense, new therapeutic strategies are necessary to reduce these effects and improve the control of infection. In this context, biogenic silver nanoparticles (AgNp-Bio) appear as a promising alternative since they have antimicrobial, antiviral, and antiparasitic activity. The purpose of this study to investigate the action of AgNp-Bio in BeWo cells, HTR-8/SVneo cells and villous explants and its effects against Toxoplasma gondii infection. Both cells and villous explants were treated with different concentrations of AgNp-Bio or combination of sulfadiazine + pyrimethamine (SDZ + PYZ) in order to verify the viability. After, cells and villi were infected and treated with AgNp-Bio or SDZ + PYZ in different concentrations to ascertain the parasite proliferation and cytokine production profile. AgNp-Bio treatment did not reduce the cell viability and villous explants. Significant reduction was observed in parasite replication in both cells and villous explants treated with silver nanoparticles and classical treatment. The AgNp-Bio treatment increased of IL-4 and IL-10 by BeWo cells, while HTR8/SVneo cells produced macrophage migration inhibitory factor (MIF) and IL-4. In the presence of T. gondii, the treatment induced high levels of MIF production by BeWo cells and IL-6 by HTR8SV/neo. In villous explants, the AgNp-Bio treatment downregulated production of IL-4, IL-6, and IL-8 after infection. In conclusion, AgNp-Bio can decrease T. gondii infection in trophoblast cells and villous explants. Therefore, this treatment demonstrated the ability to reduce the T. gondii proliferation with induction of inflammatory mediators in the cells and independent of mediators in chorionic villus which we consider the use of AgNp-Bio promising in the treatment of toxoplasmosis in BeWo and HTR8/SVneo cell models and in chorionic villi.
Collapse
Affiliation(s)
- Idessania Nazareth Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Mayara Ribeiro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Rafaela José da Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Thádia Evelyn de Araújo
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Iliana Claudia Balga Milián
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Luana Carvalho Luz
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Pâmela Mendonça Guirelli
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Gerson Nakazato
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Tiago W. P. Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
17
|
Copaifera spp. oleoresins impair Toxoplasma gondii infection in both human trophoblastic cells and human placental explants. Sci Rep 2020; 10:15158. [PMID: 32938966 PMCID: PMC7495442 DOI: 10.1038/s41598-020-72230-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022] Open
Abstract
The combination of pyrimethamine and sulfadiazine is the standard care in cases of congenital toxoplasmosis. However, therapy with these drugs is associated with severe and sometimes life-threatening side effects. The investigation of phytotherapeutic alternatives to treat parasitic diseases without acute toxicity is essential for the advancement of current therapeutic practices. The present study investigates the antiparasitic effects of oleoresins from different species of Copaifera genus against T. gondii. Oleoresins from C. reticulata, C. duckei, C. paupera, and C. pubiflora were used to treat human trophoblastic cells (BeWo cells) and human villous explants infected with T. gondii. Our results demonstrated that oleoresins were able to reduce T. gondii intracellular proliferation, adhesion, and invasion. We observed an irreversible concentration-dependent antiparasitic action in infected BeWo cells, as well as parasite cell cycle arrest in the S/M phase. The oleoresins altered the host cell environment by modulation of ROS, IL-6, and MIF production in BeWo cells. Also, Copaifera oleoresins reduced parasite replication and TNF-α release in villous explants. Anti-T. gondii effects triggered by the oleoresins are associated with immunomodulation of the host cells, as well as, direct action on parasites.
Collapse
|
18
|
Gómez-Chávez F, Cañedo-Solares I, Ortiz-Alegría LB, Flores-García Y, Figueroa-Damián R, Luna-Pastén H, Gómez-Toscano V, López-Candiani C, Arce-Estrada GE, Bonilla-Ríos CA, Mora-González JC, García-Ruiz R, Correa D. A Proinflammatory Immune Response Might Determine Toxoplasma gondii Vertical Transmission and Severity of Clinical Features in Congenitally Infected Newborns. Front Immunol 2020; 11:390. [PMID: 32231666 PMCID: PMC7082359 DOI: 10.3389/fimmu.2020.00390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/19/2020] [Indexed: 01/01/2023] Open
Abstract
Toxoplasma gondii is the etiological agent of toxoplasmosis. Mother-to-child transmission of this parasite can occur during pregnancy. Newborns with congenital toxoplasmosis may develop central nervous system impairment, with severity ranging from subclinical manifestations to death. A proinflammatory/regulated specific immune profile is crucial in the defense against the parasite; nevertheless, its role in the infected pregnant women and the congenitally infected offspring has been poorly explored, and there is still no consensus about its relation to parasite vertical transmission or to severity and dissemination in the congenitally infected newborns. This work aimed to characterize these relations by means of principal component and principal factor analyses. For this purpose, we determined the specific production of the four immunoglobulin G antibody subclasses, cytokines, and lymphocyte proliferation in the T. gondii–infected pregnant women−10 who transmitted the infection to their offspring and seven who did not—as well as in 11 newborns congenitally infected and grouped according to disease severity (five mild and six moderate/severe) and dissemination (four local and seven disseminated). We found that the immune response of nontransmitter women differed from that of the transmitters, the latter having a stronger proinflammatory response, supporting a previous report. We also found that newborns who developed moderate/severe disease presented higher levels of lymphocyte proliferation, particularly of CD8+ and CD19+ cells, a high proportion of tumor necrosis factor α producers, and reduced expression of the immune modulator transforming growth factor β, as opposed to children who developed mild clinical complications. Our results suggest that a distinctive, not regulated, proinflammatory immune response might favor T. gondii vertical transmission and the development of severe clinical manifestations in congenitally infected newborns.
Collapse
Affiliation(s)
- Fernando Gómez-Chávez
- Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico.,Cátedras CONACyT-Instituto Nacional de Pediatría, Mexico City, Mexico.,Departamento de Formación Básica Disciplinaria, ENMyH-IPN, Mexico City, Mexico
| | | | | | | | | | - Héctor Luna-Pastén
- Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| | | | | | | | | | | | - Ricardo García-Ruiz
- Laboratorio de Cannabinoides, Facultad de Medicina, UNAM, Mexico City, Mexico
| | - Dolores Correa
- Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| |
Collapse
|
19
|
Milian ICB, Silva RJ, Manzan-Martins C, Barbosa BF, Guirelli PM, Ribeiro M, de Oliveira Gomes A, Ietta F, Mineo JR, Silva Franco P, Ferro EAV. Increased Toxoplasma gondii Intracellular Proliferation in Human Extravillous Trophoblast Cells (HTR8/SVneo Line) Is Sequentially Triggered by MIF, ERK1/2, and COX-2. Front Microbiol 2019; 10:852. [PMID: 31068920 PMCID: PMC6491458 DOI: 10.3389/fmicb.2019.00852] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/02/2019] [Indexed: 12/27/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a potent pro-inflammatory cytokine, which mediates the regulation of diverse cellular functions. It is produced by extravillous trophoblastic cells and has been found to be involved in the pathogenesis of diseases caused by some protozoa, including Toxoplasma gondii. Previous studies demonstrated the ability of T. gondii to take advantage of MIF action in human trophoblast cells. However, MIF action in T. gondii-infected extravillous trophoblastic cells (HTR8/SVneo cell line) has not been fully investigated. The present study aimed to investigate the role of MIF in T. gondii-infected HTR8/SVneo cells and verify the intracellular signaling pathways triggered by this cytokine. We found that T. gondii increased MIF production by HTR8/SVneo cells, and by contrast, MIF inhibition, by ISO-1, led to a significant decrease in T. gondii proliferation and CD74 expression in HTR8/SVneo cells. Moreover, in infected HTR8/SVneo cells, the addition of recombinant MIF (rMIF) increased CD44 co-receptor expression, ERK1/2 phosphorylation, COX-2 expression, and IL-8 production, which favored T. gondii proliferation. Our findings indicate that T. gondii can use MIF to modulate important factors in HTR8/SVneo cells, being a possible explanation for the higher susceptibility of extravillous trophoblast cells than other trophoblast cell populations.
Collapse
Affiliation(s)
- Iliana Claudia Balga Milian
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Camilla Manzan-Martins
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Pamela Mendonça Guirelli
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | | | - Francesca Ietta
- Department of Life Sciences, University of Siena, Siena, Italy
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
20
|
Gómez-Chávez F, Cañedo-Solares I, Ortiz-Alegría LB, Flores-García Y, Luna-Pastén H, Figueroa-Damián R, Mora-González JC, Correa D. Maternal Immune Response During Pregnancy and Vertical Transmission in Human Toxoplasmosis. Front Immunol 2019; 10:285. [PMID: 30846989 PMCID: PMC6393384 DOI: 10.3389/fimmu.2019.00285] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 02/04/2019] [Indexed: 11/13/2022] Open
Abstract
Toxoplasmosis is a parasitic zoonosis distributed worldwide, caused by the ingestion of contaminated water/food with the parasite Toxoplasma gondii. If a pregnant woman is infected with this parasite, it may be transmitted to the fetus and produce ocular, neurological, or systemic damage with variable severity. The strength and profile of mother's immune response have been suggested as important factors involved in vertical transmission rate and severity of clinical outcome in the congenitally infected fetus. The aim of this work was to evaluate a possible relation between the mother's immune response during pregnancy and congenital transmission to the fetus. We obtained peripheral blood from T. gondii infected pregnant woman and tested it for anti T. gondii (IgG1, IgG2, IgG3, IgG4, and IgA) in serum. Peripheral blood mononuclear cells (PBMCs) were isolated to analyze the in vitro effect of soluble T. gondii antigens on proliferation and production of cytokines. We found that IgG2-4 and IgA antibodies and lymphocytes proliferation, especially CD4+, CD8+, and CD19+ were positive in a higher proportion of cases in transmitter than in non-transmitter women. Furthermore, IgG2-3 and IgA anti-Toxoplasma antibody levels were higher in those mothers who transmitted the infection than in those who did not. Interestingly, a higher proportion of positive cases to IFN-γ and negatives to the immunoregulatory cytokine TGF-β, were related to T. gondii vertical transmission. Our descriptive results are consistent with the paradoxical previous observations in murine models of congenital toxoplasmosis, which suggest that an increased immune response that protects the mothers from a disseminated or severe disease, and should protect the fetus from infection, is positively related to parasite transmission.
Collapse
Affiliation(s)
- Fernando Gómez-Chávez
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico.,Cátedras CONACyT-Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Irma Cañedo-Solares
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| | - Luz Belinda Ortiz-Alegría
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| | - Yevel Flores-García
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| | - Héctor Luna-Pastén
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| | - Ricardo Figueroa-Damián
- Servicio de infectología e Inmunología, Instituto Nacional de Perinatología, Mexico City, Mexico
| | | | - Dolores Correa
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| |
Collapse
|
21
|
Pereira ACA, Silva RJ, Franco PS, de Oliveira Gomes A, Souza G, Milian ICB, Ribeiro M, Rosini AM, Guirelli PM, Ramos ELP, Mineo TWP, Mineo JR, Silva NM, Ferro EAV, Barbosa BF. Cyclooxygenase (COX)-2 Inhibitors Reduce Toxoplasma gondii Infection and Upregulate the Pro-inflammatory Immune Response in Calomys callosus Rodents and Human Monocyte Cell Line. Front Microbiol 2019; 10:225. [PMID: 30809216 PMCID: PMC6379304 DOI: 10.3389/fmicb.2019.00225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/28/2019] [Indexed: 12/27/2022] Open
Abstract
Toxoplasma gondii is able to infect a wide range of vertebrates, including humans. Studies show that cyclooxygenase-2 (COX-2) is a modulator of immune response in multiple types of infection, such as Trypanosoma cruzi. However, the role of COX-2 during T. gondii infection is still unclear. The aim of this study was to investigate the role of COX-2 during infection by moderately or highly virulent strains of T. gondii in Calomys callosus rodents and human THP-1 cells. C. callosus were infected with 50 cysts of T. gondii (ME49), treated with COX-2 inhibitors (meloxicam or celecoxib) and evaluated to check body weight and morbidity. After 40 days, brain and serum were collected for detection of T. gondii by real-time PCR and immunohistochemistry or cytokines by CBA. Furthermore, peritoneal macrophages or THP-1 cells, infected with RH strain or uninfected, were treated with meloxicam or celecoxib to evaluate the parasite proliferation by colorimetric assay and cytokine production by ELISA. Finally, in order to verify the role of prostaglandin E2 in COX-2 mechanism, THP-1 cells were infected, treated with meloxicam or celecoxib plus PGE2, and analyzed to parasite proliferation and cytokine production. The data showed that body weight and morbidity of the animals changed after infection by T. gondii, under both treatments. Immunohistochemistry and real-time PCR showed a reduction of T. gondii in brains of animals treated with both COX-2 inhibitors. Additionally, it was observed that both COX-2 inhibitors controlled the T. gondii proliferation in peritoneal macrophages and THP-1 cells, and the treatment with PGE2 restored the parasite growth in THP-1 cells blocked to COX-2. In the serum of Calomys, upregulation of pro-inflammatory cytokines was detected, while the supernatants of peritoneal macrophages and THP-1 cells demonstrated significant production of TNF and nitrite, or TNF, nitrite and MIF, respectively, under both COX-2 inhibitors. Finally, PGE2 treatment in THP-1 cells triggered downmodulation of pro-inflammatory mediators and upregulation of IL-8 and IL-10. Thus, COX-2 is an immune mediator involved in the susceptibility to T. gondii regardless of strain or cell types, since inhibition of this enzyme induced control of infection by upregulating important pro-inflammatory mediators against Toxoplasma.
Collapse
Affiliation(s)
- Ana Carolina Alcântara Pereira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Rafaela José Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Angelica de Oliveira Gomes
- Institute of Natural and Biological Sciences, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Guilherme Souza
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Iliana Claudia Balga Milian
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Alessandra Monteiro Rosini
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Pâmela Mendonça Guirelli
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eliézer Lucas Pires Ramos
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Universidade Federal de Uberlândia, Uberlândia, Brazil
| |
Collapse
|
22
|
Franco PS, Gois PSG, de Araújo TE, da Silva RJ, de Freitas Barbosa B, de Oliveira Gomes A, Ietta F, Dos Santos LA, Dos Santos MC, Mineo JR, Ferro EAV. Brazilian strains of Toxoplasma gondii are controlled by azithromycin and modulate cytokine production in human placental explants. J Biomed Sci 2019; 26:10. [PMID: 30665403 PMCID: PMC6340180 DOI: 10.1186/s12929-019-0503-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is a protozoan parasite that causes congenital toxoplasmosis by transplacental transmission. Parasite strains are genetically diverse and disease severity is related to the genotype. In Uberlândia city, Brazil, two virulent strains were isolated: TgChBrUD1 and TgChBrUD2. Congenital toxoplasmosis is more prevalent in South America compared to Europe, and more often associated with severe symptoms, usually as a result of infection with atypical strains. METHODS Considering that T. gondii has shown high genetic diversity in Brazil, the effectiveness of traditional treatment may not be the same, as more virulent strains of atypical genotypes may predominate. Thus, the aim of this study were to evaluate the Brazilian strain infection rate in human villous explants and the azithromycin efficacy with regard to the control of these strains compared to traditional therapy. Villi were infected with RH, ME49, TgChBrUD1 or TgChBrUD2 strains and treated with azithromycin, spiramycin or a combination of pyrimethamine plus sulfadiazine. The villous viability was analyzed by LDH assay and morphological analysis. Parasite proliferation, as well as production of cytokines was analyzed by qPCR and ELISA, respectively. Statistical analysis was performed using the GraphPad Prism 5.0. RESULTS The treatments were not toxic and TgChBrUD1 infected villi showed a higher parasite burden compared with others strains. Treatments significantly reduced the intracellular proliferation of T. gondii, regardless of the strain. TgChBrUD1-infected villi produced a larger amount of MIF, IL-6 and TGF-β1 compared with other infected villi. Azithromycin treatment increased MIF production by RH- or TgChBrUD2-infected villi, but in ME49- or TgChBrUD1-infected villi, the MIF production was not altered by treatment. On the other hand, azithromycin treatment induced lower IL-6 production by ME49- or TgChBrUD1-infected villi. CONCLUSIONS Azithromycin treatment was effective against T. gondii Brazilian strains compared with conventional treatment. Also, the TgChBrUD1 strain replicated more in villi and modulated important cytokines involved in parasite control, showing that different strains use different strategies to evade the host immune response and ensure their survival.
Collapse
Affiliation(s)
- Priscila Silva Franco
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil
| | - Paula Suellen Guimarães Gois
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil
| | - Thádia Evelyn de Araújo
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil
| | - Rafaela José da Silva
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil
| | - Bellisa de Freitas Barbosa
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil
| | - Angelica de Oliveira Gomes
- Laboratório de Biologia Celular, Instituto de Ciências Biomédicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Lara Affonso Dos Santos
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil
| | - Maria Célia Dos Santos
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil
| | - José Roberto Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Av. Pará, 1720, Building: 2B, CEP, Uberlândia, 38405-320, Brazil.
| |
Collapse
|
23
|
Gomes AO, Barbosa BF, Franco PS, Ribeiro M, Silva RJ, Gois PSG, Almeida KC, Angeloni MB, Castro AS, Guirelli PM, Cândido JV, Chica JEL, Silva NM, Mineo TWP, Mineo JR, Ferro EAV. Macrophage Migration Inhibitory Factor (MIF) Prevents Maternal Death, but Contributes to Poor Fetal Outcome During Congenital Toxoplasmosis. Front Microbiol 2018; 9:906. [PMID: 29867817 PMCID: PMC5952001 DOI: 10.3389/fmicb.2018.00906] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/18/2018] [Indexed: 12/20/2022] Open
Abstract
Migration inhibitory factor (MIF) is a pro-inflammatory cytokine that plays important roles in physiology, pathology, immunology and parasitology, including the control of infection by protozoa parasites such as Toxoplasma gondii. As the MIF function in congenital toxoplasmosis is not fully elucidated yet, the present study brings new insights for T. gondii infection in the absence of MIF based on pregnant C57BL/6MIF-/- mouse models. Pregnant C57BL/6MIF-/- and C57BL/6WT mice were infected with 05 cysts of T. gondii (ME49 strain) on the first day of pregnancy (dop) and were euthanized at 8 dop. Non-pregnant and non-infected females were used as control. Our results demonstrated that MIF-/- mice have more accentuated change in body weight and succumbed to infection first than their WT counterparts. Otherwise, pregnancy outcome was less destructive in MIF-/- mice compared to WT ones, and the former had an increase in the mast cell recruitment and IDO expression and consequently presented less inflammatory cytokine production. Also, MIF receptor (CD74) was upregulated in MIF-/- mice, indicating that a compensatory mechanism may be required in this model of study. The global absence of MIF was associated with attenuation of pathology in congenital toxoplasmosis, but resulted in female death probably because of uncontrolled infection. Altogether, ours results demonstrated that part of the immune response that protects a pregnant female from T. gondii infection, favors fetal damage.
Collapse
Affiliation(s)
- Angelica O. Gomes
- Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Bellisa F. Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Priscila S. Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Rafaela J. Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Paula S. G. Gois
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Karine C. Almeida
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Mariana B. Angeloni
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Andressa S. Castro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Pâmela M. Guirelli
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - João V. Cândido
- Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Javier E. L. Chica
- Institute of Natural and Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Neide M. Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Tiago W. P. Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - José R. Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eloisa A. V. Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
24
|
da Silva RJ, Gomes AO, Franco PS, Pereira AS, Milian ICB, Ribeiro M, Fiorenzani P, dos Santos MC, Mineo JR, da Silva NM, Ferro EAV, de Freitas Barbosa B. Enrofloxacin and Toltrazuril Are Able to Reduce Toxoplasma gondii Growth in Human BeWo Trophoblastic Cells and Villous Explants from Human Third Trimester Pregnancy. Front Cell Infect Microbiol 2017; 7:340. [PMID: 28798905 PMCID: PMC5526852 DOI: 10.3389/fcimb.2017.00340] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022] Open
Abstract
Classical treatment for congenital toxoplasmosis is based on combination of sulfadiazine and pyrimethamine plus folinic acid. Due to teratogenic effects and bone marrow suppression caused by pyrimethamine, the establishment of new therapeutic strategies is indispensable to minimize the side effects and improve the control of infection. Previous studies demonstrated that enrofloxacin and toltrazuril reduced the incidence of Neospora caninum and Toxoplasma gondii infection. The aim of the present study was to evaluate the efficacy of enrofloxacin and toltrazuril in the control of T. gondii infection in human trophoblast cells (BeWo line) and in human villous explants from the third trimester. BeWo cells and villous were treated with several concentrations of enrofloxacin, toltrazuril, sulfadiazine, pyrimethamine, or combination of sulfadiazine+pyrimethamine, and the cellular or tissue viability was verified. Next, BeWo cells were infected by T. gondii (2F1 clone or the ME49 strain), whereas villous samples were only infected by the 2F1 clone. Then, infected cells and villous were treated with all antibiotics and the T. gondii intracellular proliferation as well as the cytokine production were analyzed. Finally, we evaluated the direct effect of enrofloxacin and toltrazuril in tachyzoites to verify possible changes in parasite structure. Enrofloxacin and toltrazuril did not decrease the viability of cells and villous in lower concentrations. Both drugs were able to significantly reduce the parasite intracellular proliferation in BeWo cells and villous explants when compared to untreated conditions. Regardless of the T. gondii strain, BeWo cells infected and treated with enrofloxacin or toltrazuril induced high levels of IL-6 and MIF. In villous explants, enrofloxacin induced high MIF production. Finally, the drugs increased the number of unviable parasites and triggered damage to tachyzoite structure. Taken together, it can be concluded that enrofloxacin and toltrazuril are able to control T. gondii infection in BeWo cells and villous explants, probably by a direct action on the host cells and parasites, which leads to modifications of cytokine release and tachyzoite structure.
Collapse
Affiliation(s)
- Rafaela J. da Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| | - Angelica O. Gomes
- Departament of Morphology, Federal University of Triângulo MineiroUberaba, Brazil
| | - Priscila S. Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| | - Ariane S. Pereira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| | - Iliana C. B. Milian
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| | - Mayara Ribeiro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| | - Paolo Fiorenzani
- Department of Medical, Surgery and Neuroscience, University of SienaSiena, Italy
| | - Maria C. dos Santos
- Department of Gynecology and Obstetrics, Faculty of Medicine, Federal University of UberlândiaUberlândia, Brazil
| | - José R. Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| | - Neide M. da Silva
- Laboratory of Immunopathology, Institute of Biomedical Science, Federal University of UberlandiaUberlândia, Brazil
| | - Eloisa A. V. Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| | - Bellisa de Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Science, Federal University of UberlândiaUberlândia, Brazil
| |
Collapse
|
25
|
Rottlerin-mediated inhibition of Toxoplasma gondii growth in BeWo trophoblast-like cells. Sci Rep 2017; 7:1279. [PMID: 28455500 PMCID: PMC5430667 DOI: 10.1038/s41598-017-01525-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/29/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a crucial and physiological process for cell survival from yeast to mammals, including protozoan parasites. Toxoplasma gondii, an intracellular parasite, typically exploits autophagic machinery of host cell; however host cell upregulates autophagy to combat the infection. Herein we tested the efficacy of Rottlerin, a natural polyphenol with autophagic promoting properties, against Toxoplasma infection on the chorioncarcinoma-derived cell line BeWo. We found that Rottlerin, at sub-toxic doses, induced morphological and biochemical alterations associated with autophagy and decreased Toxoplasma growth in infected cells. Although autophagy was synergically promoted by Toxoplasma infection in combination with Rottlerin treatment, the use of the autophagy inhibitor chloroquine revealed that Rottlerin anti-parasitic effect was largely autophagy-independent and likely mediated by the converging inhibitory effect of Rottlerin and Toxoplasma in host protein translation, mediated by mTOR inhibition and eIF2α phosphorylation. Both events, which on one hand could explain the additive effect on autophagy induction, on the other hand led to inhibition of protein synthesis, thereby depriving Toxoplasma of metabolically essential components for multiplication. We suggest that modulation of the competition between pathogen requirement and host cell defense might be an attractive, novel therapeutic approach against Toxoplasma infection and encourage the development of Rottlerin-based new therapeutic formulations.
Collapse
|
26
|
Przybyl L, Haase N, Golic M, Rugor J, Solano ME, Arck PC, Gauster M, Huppertz B, Emontzpohl C, Stoppe C, Bernhagen J, Leng L, Bucala R, Schulz H, Heuser A, Weedon-Fekjær MS, Johnsen GM, Peetz D, Luft FC, Staff AC, Müller DN, Dechend R, Herse F. CD74-Downregulation of Placental Macrophage-Trophoblastic Interactions in Preeclampsia. Circ Res 2016; 119:55-68. [PMID: 27199465 DOI: 10.1161/circresaha.116.308304] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/19/2016] [Indexed: 01/28/2023]
Abstract
RATIONALE We hypothesized that cluster of differentiation 74 (CD74) downregulation on placental macrophages, leading to altered macrophage-trophoblast interaction, is involved in preeclampsia. OBJECTIVE Preeclamptic pregnancies feature hypertension, proteinuria, and placental anomalies. Feto-placental macrophages regulate villous trophoblast differentiation during placental development. Disturbance of this well-balanced regulation can lead to pathological pregnancies. METHODS AND RESULTS We performed whole-genome expression analysis of placental tissue. CD74 was one of the most downregulated genes in placentas from preeclamptic women. By reverse transcriptase-polymerase chain reaction, we confirmed this finding in early-onset (<34 gestational week, n=26) and late-onset (≥34 gestational week, n=24) samples from preeclamptic women, compared with healthy pregnant controls (n=28). CD74 protein levels were analyzed by Western blot and flow cytometry. We identified placental macrophages to express CD74 by immunofluorescence, flow cytometry, and RT-PCR. CD74-positive macrophages were significantly reduced in preeclamptic placentas compared with controls. CD74-silenced macrophages showed that the adhesion molecules ALCAM, ICAM4, and Syndecan-2, as well as macrophage adhesion to trophoblasts were diminished. Naive and activated macrophages lacking CD74 showed a shift toward a proinflammatory signature with an increased secretion of tumor necrosis factor-α, chemokine (C-C motif) ligand 5, and monocyte chemotactic protein-1, when cocultured with trophoblasts compared with control macrophages. Trophoblasts stimulated by these factors express more CYP2J2, sFlt1, TNFα, and IL-8. CD74-knockout mice showed disturbed placental morphology, reduced junctional zone, smaller placentas, and impaired spiral artery remodeling with fetal growth restriction. CONCLUSIONS CD74 downregulation in placental macrophages is present in preeclampsia. CD74 downregulation leads to altered macrophage activation toward a proinflammatory signature and a disturbed crosstalk with trophoblasts.
Collapse
Affiliation(s)
- Lukasz Przybyl
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Nadine Haase
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Michaela Golic
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Julianna Rugor
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Maria Emilia Solano
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Petra Clara Arck
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Martin Gauster
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Berthold Huppertz
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Christoph Emontzpohl
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Christian Stoppe
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Jürgen Bernhagen
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Lin Leng
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Richard Bucala
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Herbert Schulz
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Arnd Heuser
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - M Susanne Weedon-Fekjær
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Guro M Johnsen
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Dirk Peetz
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Friedrich C Luft
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Anne Cathrine Staff
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Dominik N Müller
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Ralf Dechend
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.)
| | - Florian Herse
- From the Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Medical Faculty, Berlin, Germany (L.P., N.H., M. Golic, J.R., F.C.L., D.N.M., R.D., F.H.); Berlin Institute of Health (BIH), Berlin, Germany (L.P., N.H., M. Golic, J.R., D.N.M., R.D., F.H.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (N.H., H.S., A.H., F.C.L., D.N.M., F.H.); Departments of Obstetrics, Gynecology, and Senology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany (M. Golic); Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.E.S., P.C.A.); Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria (M. Gauster, B.H.); Institute of Biochemistry and Molecular Cell Biology (C.E., C.S., J.B.) and Department of Anesthesiology (C.S.), RWTH Aachen University, Aachen, Germany; Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany (J.B.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (J.B.); Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (L.L., R.B.); Cologne Center for Genomics (CCG), University of Cologne, Köln, Germany (H.S.); Departments of Obstetrics and Gynaecology, Oslo University Hospital, Ulleval, Norway (M.S.W.-F., G.M.J., A.C.S.); University of Oslo, Oslo, Norway (M.S.W.-F., G.M.J., A.C.S.); and HELIOS-Klinikum, Berlin, Germany (D.P., R.D.).
| |
Collapse
|
27
|
Is Toxoplasma gondii type related to clinical outcome in human congenital infection? Systematic and critical review. Eur J Clin Microbiol Infect Dis 2016; 35:1079-88. [PMID: 27146878 DOI: 10.1007/s10096-016-2656-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/19/2016] [Indexed: 01/23/2023]
Abstract
In human congenital toxoplasmosis the effects of parasite burden and pregnancy time at infection on clinical outcome are well known, but there is controversy regarding the role of Toxoplasma gondii type. Through a systematic review of the literature, we aimed to discern if T. gondii type has a role on clinical outcome in human congenital toxoplasmosis. We built up a database of congenital toxoplasmosis from reports of cases, case series and screening-based cohorts, which had information about parasite type, gestation time at maternal infection and/or clinical outcome in the product. Then, we obtained frequencies for loci used to genotype geographical origin of cases and types found. Also, odds ratios were calculated for association between time of maternal infection or parasite type on outcome. Type II parasites were the most common in Europe, Asia and Africa, while in America there were mainly atypical strains. More newborns with clinical problems were born from mothers infected during the first half of gestation than from those acquiring the parasite after week 24, regardless of parasite genotype (92.9 vs. 16.1 %, OR = 67.9, CI95 25.4-181.6). Type I and atypical parasites were associated with clinical problems as opposed to types II and III, regardless of pregnancy period at infection (86.9 vs. 72.9 %, OR = 2.47, CI95 1.1-5.4). A significant and remarkable tendency of type I parasites to be present during early pregnancy was also observed (94.4 vs. 5.6 %, P < 0.009). In addition to parasite burden and period of gestation, T. gondii genotype seems involved in CT clinical outcome.
Collapse
|
28
|
Costa AF, Gomes SZ, Lorenzon-Ojea AR, Martucci M, Faria MR, Pinto DDS, Oliveira SF, Ietta F, Paulesu L, Bevilacqua E. Macrophage migration inhibitory factor induces phosphorylation of Mdm2 mediated by phosphatidylinositol 3-kinase/Akt kinase: Role of this pathway in decidual cell survival. Placenta 2016; 41:27-38. [PMID: 27208405 DOI: 10.1016/j.placenta.2016.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 01/10/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway has an anti-apoptotic effect through several downstream targets, which includes activation of the transformed mouse 3T3 cell double-minute 2 (Mdm2) protein, its translocation to the nucleus and degradation of the tumor suppressor p53. We show that Mif, the Macrophage Migration Inhibitory Factor, an important cytokine at the maternal fetal interface in several species, triggers phosphorylation of Mdm2 protein in a PI3K/Akt-dependent manner, thereby preventing apoptosis in cultured mouse decidual cells. Inhibition of Akt and PI3K suppresses the pathway. Mif treatment also changes the nuclear translocation of p53 and interferes with the apoptotic fate of these cells when challenged with reactive oxygen species. In conclusion, an important mechanism has been found underlying decidual cell survival through Akt signaling pathway activated by Mif, suggesting a role for this cytokine in decidual homeostasis and in the integrity of the maternal-fetal barrier that is essential for successful gestation.
Collapse
Affiliation(s)
- Adriana Fraga Costa
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Stomatology, Dental School, University of São Paulo, São Paulo, Brazil
| | - Sara Zago Gomes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline R Lorenzon-Ojea
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariane Martucci
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Miriam Rubio Faria
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Sergio F Oliveira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Luana Paulesu
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Estela Bevilacqua
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
29
|
Harker KS, Ueno N, Lodoen MB. Toxoplasma gondiidissemination: a parasite's journey through the infected host. Parasite Immunol 2015; 37:141-9. [DOI: 10.1111/pim.12163] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 11/11/2014] [Indexed: 12/30/2022]
Affiliation(s)
- K. S. Harker
- Department of Molecular Biology and Biochemistry and the Institute for Immunology; University of California; Irvine CA USA
| | - N. Ueno
- Department of Molecular Biology and Biochemistry and the Institute for Immunology; University of California; Irvine CA USA
| | - M. B. Lodoen
- Department of Molecular Biology and Biochemistry and the Institute for Immunology; University of California; Irvine CA USA
| |
Collapse
|
30
|
Barbosa BF, Lopes-Maria JB, Gomes AO, Angeloni MB, Castro AS, Franco PS, Fermino ML, Roque-Barreira MC, Ietta F, Martins-Filho OA, Silva DAO, Mineo JR, Ferro EAV. IL10, TGF beta1, and IFN gamma modulate intracellular signaling pathways and cytokine production to control Toxoplasma gondii infection in BeWo trophoblast cells. Biol Reprod 2015; 92:82. [PMID: 25673564 DOI: 10.1095/biolreprod.114.124115] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Considering that interleukin 10 (IL10), transforming growth factor beta1 (TGFB1), and interferon gamma (IFNG) are involved in the susceptibility of BeWo trophoblast cells to Toxoplasma gondii infection, the aim of the present study was to investigate the effector mechanisms triggered by these cytokines in the control of T. gondii in BeWo cells. For this purpose, infected/uninfected BeWo cells were treated with IL10, TGFB1 (50 ng/ml), and IFNG (20 or 100 ng/ml) in order to verify the phosphorylation of signal transducers and activators of transcription 1 (STAT1), STAT3, and Smad2, parasite intracellular proliferation, as well as the Th1/Th2/IL17A cytokine production. The treatment of BeWo cells with IL10 and TGFB1 favored T. gondii proliferation, and these findings were associated with STAT3 and Smad2 phosphorylation, respectively (P < 0.05). Also, these cytokine treatments were able to down-modulate TNF alpha (TNFA) and IL6 production (P < 0.05). Low concentration of IFNG was unable to control T. gondii infection but was able to trigger STAT1 phosphorylation and up-regulate IL6 and IL17A production; whereas a high concentration of IFNG was unable to activate STAT1 but down-modulated IL6 and TNFA and increased T. gondii proliferation (P < 0.05). IL10, TGFB1, and IFNG regulate a differential T. gondii proliferation in BeWo cells because they distinctly trigger intracellular signaling pathways and cytokine production, especially IL6 and TNFA. Our data open new windows to understand the mechanisms triggered by IL10, TGFB1, and IFNG at the maternal-fetal interface in the presence of T. gondii, contributing to recognizing the importance of these effector mechanisms involved in the vertical transmission of this parasite.
Collapse
Affiliation(s)
- Bellisa Freitas Barbosa
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Janice Buiate Lopes-Maria
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Angelica Oliveira Gomes
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Mariana Bodini Angeloni
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Andressa Silva Castro
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Priscila Silva Franco
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Marise Lopes Fermino
- Department of Cellular and Molecular Biology, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cellular and Molecular Biology, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Francesca Ietta
- Department of Life Science, University of Siena, Siena, Italy
| | - Olindo Assis Martins-Filho
- Laboratory of Chagas Disease, René Rachou Research Center, Fiocruz, Belo Horizonte, Minas Gerais, Brazil
| | - Deise Aparecida Oliveira Silva
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
31
|
Barbosa B, Paulesu L, Ietta F, Bechi N, Romagnoli R, Gomes A, Favoreto-Junior S, Silva D, Mineo J, Mineo T, Ferro E. Susceptibility to Toxoplasma gondii proliferation in BeWo human trophoblast cells is dose-dependent of macrophage migration inhibitory factor (MIF), via ERK1/2 phosphorylation and prostaglandin E2 production. Placenta 2014; 35:152-62. [DOI: 10.1016/j.placenta.2013.12.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 12/19/2013] [Accepted: 12/24/2013] [Indexed: 12/21/2022]
|
32
|
Bevilacqua E, Paulesu L, Ferro EAV, Ietta F, Faria MR, Lorenzon AR, Costa AF, Martucci M. Review: putative roles for the macrophage migratory inhibitory factor at the maternal fetal interface. Placenta 2013; 35 Suppl:S51-6. [PMID: 24215782 DOI: 10.1016/j.placenta.2013.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 10/22/2013] [Accepted: 10/23/2013] [Indexed: 12/22/2022]
Abstract
Complex and dynamic networks of molecules participate in the essential interactions between maternal organism, placenta and fetus in a healthy and successful pregnancy. Macrophage migratory inhibitory factor (MIF) is one of several molecules produced at implantation sites; MIF is mostly expressed by trophoblast cells. This has led to expectations of MIF's relevance as a partner in the maternal/fetal dialog. MIF is known by its biological interactions and functional roles as an activator of innate immunity, regulating subsequent adaptive responses, which include inhibition of migration of mononuclear cells in vitro, antagonism of glucocorticoids, and regulation of expression of Toll-like receptor 4. Beyond roles in the inflammatory response, MIF can interfere with proliferative activities in different cell types, as well as with cell death pathways. This intriguing factor found at the human, porcine, ovine, bovine and rodent maternal-fetal interfaces is present in a time- and spatially-dependent manner, indicating regulatory roles in the process of embryo implantation, placental development, maintenance of pregnancy and birth. Here, we will review MIF participation in placental physiology, including new evidence for a dialog with uterine cells, and a potential role in protection of uterine decidual cells.
Collapse
Affiliation(s)
- E Bevilacqua
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil.
| | - L Paulesu
- Department of Life Sciences, Reproductive Physiology, University of Siena, Siena, Italy
| | - E A V Ferro
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlandia, MG, Brazil
| | - F Ietta
- Department of Life Sciences, Reproductive Physiology, University of Siena, Siena, Italy
| | - M R Faria
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - A R Lorenzon
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - A F Costa
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil; Laboratory of Cell Biology, Stomatology Department, Dentistry School, University of São Paulo, SP, Brazil
| | - M Martucci
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| |
Collapse
|
33
|
Põlajeva J, Bergström T, Edqvist PH, Lundequist A, Sjösten A, Nilsson G, Smits A, Bergqvist M, Pontén F, Westermark B, Pejler G, Forsberg Nilsson K, Tchougounova E. Glioma-derived macrophage migration inhibitory factor (MIF) promotes mast cell recruitment in a STAT5-dependent manner. Mol Oncol 2013; 8:50-8. [PMID: 24091309 DOI: 10.1016/j.molonc.2013.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 08/27/2013] [Accepted: 09/09/2013] [Indexed: 11/15/2022] Open
Abstract
Recently, glioma research has increased its focus on the diverse types of cells present in brain tumors. We observed previously that gliomas are associated with a profound accumulation of mast cells (MCs) and here we investigate the underlying mechanism. Gliomas express a plethora of chemoattractants. First, we demonstrated pronounced migration of human MCs toward conditioned medium from cultures of glioma cell lines. Subsequent cytokine array analyses of media from cells, cultured in either serum-containing or -free conditions, revealed a number of candidates which were secreted in high amounts in both cell lines. Among these, we then focused on macrophage migration inhibitory factor (MIF), which has been reported to be pro-inflammatory and -tumorigenic. Infiltration of MCs was attenuated by antibodies that neutralized MIF. Moreover, a positive correlation between the number of MCs and the level of MIF in a large cohort of human glioma tissue samples was observed. Further, both glioma-conditioned media and purified MIF promoted differential phosphorylation of a number of signaling molecules, including signal transducer and activator of transcription 5 (STAT5), in MCs. Inhibition of pSTAT5 signaling significantly attenuated the migration of MCs toward glioma cell-conditioned medium shown to contain MIF. In addition, analysis of tissue microarrays (TMAs) of high-grade gliomas revealed a direct correlation between the level of pSTAT5 in MCs and the level of MIF in the medium. In conclusion, these findings indicate the important influence of signaling cascades involving MIF and STAT5 on the recruitment of MCs to gliomas.
Collapse
Affiliation(s)
- Jelena Põlajeva
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden.
| | - Tobias Bergström
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden.
| | - Per-Henrik Edqvist
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden.
| | - Anders Lundequist
- Swedish University of Agricultural Sciences, Department of Anatomy, Physiology and Biochemistry, BMC, Box 575, SE-751 23 Uppsala, Sweden.
| | - Anna Sjösten
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden
| | - Gunnar Nilsson
- Karolinska Institutet, Department of Medicine, Karolinska University Hospital Solna, SE-171 76 Stockholm, Sweden.
| | - Anja Smits
- Uppsala University Hospital, Department of Neuroscience, Neurology, SE-751 85 Uppsala, Sweden.
| | - Michael Bergqvist
- Uppsala University Hospital, Department of Neuroscience, Neurology, SE-751 85 Uppsala, Sweden.
| | - Fredrik Pontén
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden.
| | - Bengt Westermark
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden.
| | - Gunnar Pejler
- Swedish University of Agricultural Sciences, Department of Anatomy, Physiology and Biochemistry, BMC, Box 575, SE-751 23 Uppsala, Sweden.
| | - Karin Forsberg Nilsson
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden.
| | - Elena Tchougounova
- Uppsala University, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden.
| |
Collapse
|
34
|
Castro AS, Alves CMOS, Angeloni MB, Gomes AO, Barbosa BF, Franco PS, Silva DAO, Martins-Filho OA, Mineo JR, Mineo TWP, Ferro EAV. Trophoblast cells are able to regulate monocyte activity to control Toxoplasma gondii infection. Placenta 2013; 34:240-7. [PMID: 23294571 DOI: 10.1016/j.placenta.2012.12.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/30/2012] [Accepted: 12/12/2012] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Toxoplasma gondii is an intracellular parasite that causes severe disease when the infection occurs during pregnancy. Trophoblast cells constitute an important maternal-fetal barrier, with monocytes concentrating around them. Thus, interactions between trophoblasts and monocytes are important for maintaining a successful pregnancy, especially in cases of infection. This study aimed to evaluate the role of trophoblast cells (BeWo line) on monocyte (THP-1 line) activity in the presence or absence of T. gondii infection. METHODS THP-1 cells were stimulated with supernatants of BeWo cells, previously infected or not with T. gondii, and then infected with parasites. The supernatant of both cells were collected and analyzed for cytokine production and T. gondii proliferation in THP-1 cells was determined. RESULTS The results showed that after infection, the pattern of cytokines secreted by THP-1 and BeWo cells was characterized as a pro-inflammatory profile. Furthermore, supernatant of BeWo cells infected or not, was able to change the cytokine profile secreted by infected THP-1 cells, and this supernatant became THP-1 cells more able to control T. gondii proliferation than those that had not been stimulated. DISCUSSION This effect was associated with secretion of interleukin (IL)-6 by the THP-1 cells and soluble factors secreted by BeWo cells, such as IL-6 and MIF. CONCLUSION Together, these results suggest that trophoblast cells are able to modulate monocyte activity, resulting in the control of T. gondii infection and subsequent maintenance of pregnancy.
Collapse
Affiliation(s)
- A S Castro
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, Av. Pará, 1720, 38405-320 Uberlândia, MG, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Macrophage migration inhibitory factor in protozoan infections. J Parasitol Res 2012; 2012:413052. [PMID: 22496958 PMCID: PMC3306950 DOI: 10.1155/2012/413052] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 11/01/2011] [Accepted: 11/07/2011] [Indexed: 12/12/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine that plays a central role in immune and inflammatory responses. In the present paper, we discussed the participation of MIF in the immune response to protozoan parasite infections. As a general trend, MIF participates in the control of parasite burden at the expense of promoting tissue damage due to increased inflammation.
Collapse
|
36
|
Carlier Y, Truyens C, Deloron P, Peyron F. Congenital parasitic infections: a review. Acta Trop 2012; 121:55-70. [PMID: 22085916 DOI: 10.1016/j.actatropica.2011.10.018] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 10/27/2011] [Accepted: 10/29/2011] [Indexed: 12/11/2022]
Abstract
This review defines the concepts of maternal-fetal (congenital) and vertical transmissions (mother-to-child) of pathogens and specifies the human parasites susceptible to be congenitally transferred. It highlights the epidemiological features of this transmission mode for the three main congenital parasitic infections due to Toxoplasma gondii, Trypanosoma cruzi and Plasmodium sp. Information on the possible maternal-fetal routes of transmission, the placental responses to infection and timing of parasite transmission are synthesized and compared. The factors susceptible to be involved in parasite transmission and development of congenital parasitic diseases, such as the parasite genotypes, the maternal co-infections and parasitic load, the immunological features of pregnant women and the capacity of some fetuses/neonates to overcome their immunological immaturity to mount an immune response against the transmitted parasites are also discussed and compared. Analysis of clinical data indicates that parasitic congenital infections are often asymptomatic, whereas symptomatic newborns generally display non-specific symptoms. The long-term consequences of congenital infections are also mentioned, such as the imprinting of neonatal immune system and the possible trans-generational transmission. The detection of infection in pregnant women is mainly based on standard serological or parasitological investigations. Amniocentesis and cordocentesis can be used for the detection of some fetal infections. The neonatal infection can be assessed using parasitological, molecular or immunological methods; the place of PCR in such neonatal diagnosis is discussed. When such laboratory diagnosis is not possible at birth or in the first weeks of life, standard serological investigations can also be performed 8-10 months after birth, to avoid detection of maternal transmitted antibodies. The specific aspects of treatment of T. gondii, T. cruzi and Plasmodium congenital infections are mentioned. The possibilities of primary and secondary prophylaxes, as well as the available WHO corresponding recommendations are also presented.
Collapse
|
37
|
Abstract
Toxoplasma gondii is a ubiquitous, obligate intracellular parasite capable of crossing the placenta to cause spontaneous abortion, preterm labor, or significant disease in the surviving neonate. Exploration of the cellular and histological components of the placental barrier is in its infancy, and both how and where T. gondii breaches it are unknown. The human placenta presents two anatomical interfaces between maternal cells and fetal cells (trophoblasts): (i) the villous region where maternal blood bathes syncytialized trophoblasts for nutrient exchange and (ii) the maternal decidua, where mononuclear, extravillous trophoblasts anchor the villous region to the uterus. Using first-trimester human placental explants, we demonstrate that the latter site is significantly more vulnerable to infection, despite presenting a vastly smaller surface. This is consistent with past findings concerning two vertically transmitted viruses and one bacterium. We further explore whether three genetically distinct T. gondii types (I, II, and III) are capable of preferential placental infection and survival in this model. We find no difference in these strains' ability to infect placental explants; however, slightly slower growth is evident in type II (Prugniaud [Pru]) parasites relative to other cell types, although this did not quite achieve statistical significance.
Collapse
|
38
|
de Dios Rosado J, Rodriguez-Sosa M. Macrophage migration inhibitory factor (MIF): a key player in protozoan infections. Int J Biol Sci 2011; 7:1239-56. [PMID: 22110378 PMCID: PMC3221362 DOI: 10.7150/ijbs.7.1239] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 12/27/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine produced by the pituitary gland and multiple cell types, including macrophages (Mø), dendritic cells (DC) and T-cells. Upon releases MIF modulates the expression of several inflammatory molecules, such as TNF-α, nitric oxide and cyclooxygenase 2 (COX-2). These important MIF characteristics have prompted investigators to study its role in parasite infections. Several reports have demonstrated that MIF plays either a protective or deleterious role in the immune response to different pathogens. Here, we review the role of MIF in the host defense response to some important protozoan infections.
Collapse
Affiliation(s)
| | - Miriam Rodriguez-Sosa
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), 54090 Tlalnepantla, Estado de México, México
| |
Collapse
|
39
|
Macrophage migration inhibitory factor in fetoplacental tissues from preeclamptic pregnancies with or without fetal growth restriction. Clin Dev Immunol 2011; 2012:639342. [PMID: 22007254 PMCID: PMC3189467 DOI: 10.1155/2012/639342] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/02/2011] [Accepted: 08/03/2011] [Indexed: 12/17/2022]
Abstract
The proinflammatory cytokine MIF (macrophage migration inhibitory factor) is involved in physiological and pathological processes in pregnancy. MIF maternal serum levels are increased in preeclampsia (PE). We hypothesize that pregnancy tissues are the source of MIF overexpression in PE. MIF protein was studied in maternal sera, placental tissues, fetal membranes, and umbilical cord of 8 control and 20 PE pregnancies: 10 with normal fetal growth (PE-AGA) and 10 with fetal growth restriction (PE-FGR). MIF levels were significantly higher in PE-AGA membranes than in controls and PE-FGR. In PE-FGR, MIF cord concentrations were higher than in PE-AGA while MIF placental levels were lower than in controls. MIF maternal serum levels were higher in PE, compared to controls, and the difference was mainly due to PE-FGR samples. These data support MIF involvement in PE pathogenesis and suggest that different pregnancy tissues contribute to MIF production in PE with and without fetoplacental compromise.
Collapse
|
40
|
Effect of macrophage migration inhibitory factor (MIF) in human placental explants infected with Toxoplasma gondii depends on gestational age. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2792-801. [PMID: 21641401 DOI: 10.1016/j.ajpath.2011.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 02/22/2011] [Accepted: 02/28/2011] [Indexed: 12/13/2022]
Abstract
Because macrophage migration inhibitory factor (MIF) is a key cytokine in pregnancy and has a role in inflammatory response and pathogen defense, the objective of the present study was to investigate the effects of MIF in first- and third-trimester human placental explants infected with Toxoplasma gondii. Explants were treated with recombinant MIF, IL-12, interferon-γ, transforming growth factor-β1, or IL-10, followed by infection with T. gondii RH strain tachyzoites. Supernatants of cultured explants were assessed for MIF production. Explants were processed for morphologic analysis, immunohistochemistry, and real-time PCR analysis. Comparison of infected and stimulated explants versus noninfected control explants demonstrated a significant increase in MIF release in first-trimester but not third-trimester explants. Tissue parasitism was higher in third- than in first-trimester explants. Moreover, T. gondii DNA content was lower in first-trimester explants treated with MIF compared with untreated explants. However, in third-trimester explants, MIF stimulus decreased T. gondii DNA content only at the highest concentration of the cytokine. In addition, high expression of MIF receptor was observed in first-trimester placental explants, whereas MIF receptor expression was low in third-trimester explants. In conclusion, MIF was up-regulated and demonstrated to be important for control of T. gondii infection in first-trimester explants, whereas lack of MIF up-regulation in third-trimester placentas may be involved in higher susceptibility to infection at this gestational age.
Collapse
|
41
|
Cavalcanti MG, Mesquita JS, Madi K, Feijó DF, Assunção-Miranda I, Souza HSP, Bozza MT. MIF participates in Toxoplasma gondii-induced pathology following oral infection. PLoS One 2011; 6:e25259. [PMID: 21977228 PMCID: PMC3178626 DOI: 10.1371/journal.pone.0025259] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 08/30/2011] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is essential for controlling parasite burden and survival in a model of systemic Toxoplasma gondii infection. Peroral T. gondii infection induces small intestine necrosis and death in susceptible hosts, and in many aspects resembles inflammatory bowel disease (IBD). Considering the critical role of MIF in the pathogenesis of IBD, we hypothesized that MIF participates in the inflammatory response induced by oral infection with T. gondii. METHODOLOGY/PRINCIPAL FINDINGS Mif deficient (Mif(-/-)) and wild-type mice in the C57Bl/6 background were orally infected with T. gondii strain ME49. Mif(-/-) mice had reduced lethality, ileal inflammation and tissue damage despite of an increased intestinal parasite load compared to wt mice. Lack of MIF caused a reduction of TNF-α, IL-12, IFN-γ and IL-23 and an increased expression of IL-22 in ileal mucosa. Moreover, suppressed pro-inflammatory responses at the ileal mucosa observed in Mif(-/-) mice was not due to upregulation of IL-4, IL-10 or TGF-β. MIF also affected the expression of matrix metalloproteinase-9 (MMP-9) but not MMP-2 in the intestine of infected mice. Signs of systemic inflammation including the increased concentrations of inflammatory cytokines in the plasma and liver damage were less pronounced in Mif(-/-) mice compared to wild-type mice. CONCLUSION/SIGNIFICANCE In conclusion, our data suggested that in susceptible hosts MIF controls T. gondii infection with the cost of increasing local and systemic inflammation, tissue damage and death.
Collapse
Affiliation(s)
- Marta G. Cavalcanti
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Serviço de Doenças Infecciosas e Parasitárias, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Jacilene S. Mesquita
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Kalil Madi
- Departamento de Patologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório Sérgio Franco, Rio de Janeiro, Brazil
| | - Daniel F. Feijó
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Iranaia Assunção-Miranda
- Departamento de Virologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Heitor S. P. Souza
- Departamento de Clínica Médica, Laboratório Multidisciplinar de Pesquisa, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Departamento de Clínica Médica, Laboratório Multidisciplinar de Pesquisa, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Franco PS, Gomes AO, Barbosa BF, Angeloni MB, Silva NM, Teixeira-Carvalho A, Martins-Filho OA, Silva DAO, Mineo JR, Ferro EAV. Azithromycin and spiramycin induce anti-inflammatory response in human trophoblastic (BeWo) cells infected by Toxoplasma gondii but are able to control infection. Placenta 2011; 32:838-44. [PMID: 21908042 DOI: 10.1016/j.placenta.2011.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 08/19/2011] [Accepted: 08/24/2011] [Indexed: 10/17/2022]
Abstract
Toxoplasma gondii is an important pathogen which may cause fetal infection if primary infection. Our previous studies have used human choriocarcinoma trophoblastic cells (BeWo cell line) as experimental model of T. gondii infection involving placental microenvironment. This study aimed to examine the effects of azithromycin and spiramycin against T. gondii infection in BeWo cells. Cells were treated with different concentrations of the macrolide antibiotics and analyzed first for cell viability using thiazolyl blue tetrazole (MTT) assay. As cell viability was significantly decreased with drug concentrations higher than 400 μg/mL, the concentration range used in further experiments was from 50 to 400 μg/mL. The number of infected cells and intracellular replication of T. gondii decreased after treatment with each drug. The infection induced up-regulation of the macrophage migration inhibitory factor (MIF), which was also enhanced in infected cells after treatment with azithromycin, but not with spiramycin. Analysis of the cytokine profile showed increase TNF-α, IL-10 and IL-4 production, but decreased IFN-γ levels, were detected in infected cells and treated with each drug. In conclusion, treatment of human trophoblastic BeWo cells with with azithromycin or spiramycin is able to control the infection and replication of T. gondii. In addition, treatment with these macrolides, especially with azityromycin induces an anti-inflammatory response and high MIF production, which can be important for the establishment and maintenance of a viable pregnancy during T. gondii infection.
Collapse
Affiliation(s)
- P S Franco
- Laboratory of Histology and Embryology, Institute of Biomedical Sciences, Federal University of Uberlândia, 38405-320 Uberlândia, MG, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Shyu LY, Yeh TM, Chang HH, Lin DPC, Teng YH, Chen LC, Lee HH. Macrophage migration inhibitory factor induces ICAM-1and thrombomobulin expression in vitro. Thromb Res 2011; 129:43-9. [PMID: 21890178 DOI: 10.1016/j.thromres.2011.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 07/20/2011] [Accepted: 08/08/2011] [Indexed: 11/25/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an important cytokine in the modulation of inflammatory and immune responses, but its role in coagulation remains to be elucidated. In this study, we investigated the potential role of MIF in coagulation through its influence on two factors, thrombomodulin (TM) and intercellular adhesion molecule-1 (ICAM-1). Recombinant human MIF was added to human microvascular endothelial cell line (HMEC-1) to investigate its influence on the expression of TM and ICAM-1. The results showed that both TM and ICAM-1 were induced with MIF addition in a dose-dependent and time-dependent manner. The expression of ICAM-1 and TM was increased as MIF doses were increased, with the highest expression seen at 12 hr after 400 ng/ml of MIF treatment. Besides, anti-MIF antibody treatment reduced the TM expression in HMEC-1 cells. In conclusion, our data support a role of MIF as an important factor in the regulation of coagulation.
Collapse
Affiliation(s)
- Ling-Yuh Shyu
- Institute of Medicine, Chung Shan Medical University, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
44
|
Coxiella burnetii, the agent of Q fever, replicates within trophoblasts and induces a unique transcriptional response. PLoS One 2010; 5:e15315. [PMID: 21179488 PMCID: PMC3001886 DOI: 10.1371/journal.pone.0015315] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 11/08/2010] [Indexed: 01/27/2023] Open
Abstract
Q fever is a zoonosis caused by Coxiella burnetii, an obligate intracellular bacterium typically found in myeloid cells. The infection is a source of severe obstetrical complications in humans and cattle and can undergo chronic evolution in a minority of pregnant women. Because C. burnetii is found in the placentas of aborted fetuses, we investigated the possibility that it could infect trophoblasts. Here, we show that C. burnetii infected and replicated in BeWo trophoblasts within phagolysosomes. Using pangenomic microarrays, we found that C. burnetii induced a specific transcriptomic program. This program was associated with the modulation of inflammatory responses that were shared with inflammatory agonists, such as TNF, and more specific responses involving genes related to pregnancy development, including EGR-1 and NDGR1. In addition, C. burnetii stimulated gene networks organized around the IL-6 and IL-13 pathways, which both modulate STAT3. Taken together, these results revealed that trophoblasts represent a protective niche for C. burnetii. The activation program induced by C. burnetii in trophoblasts may allow bacterial replication but seems unable to interfere with the development of normal pregnancy. Such pathophysiologocal processes should require the activation of immune placental cells associated with trophoblasts.
Collapse
|
45
|
Silva NM, Manzan RM, Carneiro WP, Milanezi CM, Silva JS, Ferro EAV, Mineo JR. Toxoplasma gondii: The severity of toxoplasmic encephalitis in C57BL/6 mice is associated with increased ALCAM and VCAM-1 expression in the central nervous system and higher blood–brain barrier permeability. Exp Parasitol 2010; 126:167-77. [DOI: 10.1016/j.exppara.2010.04.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 03/15/2010] [Accepted: 04/20/2010] [Indexed: 01/09/2023]
|
46
|
Carvalho JV, Alves CMOS, Cardoso MRD, Mota CM, Barbosa BF, Ferro EAV, Silva NM, Mineo TWP, Mineo JR, Silva DAO. Differential susceptibility of human trophoblastic (BeWo) and uterine cervical (HeLa) cells to Neospora caninum infection. Int J Parasitol 2010; 40:1629-37. [PMID: 20708622 DOI: 10.1016/j.ijpara.2010.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 06/22/2010] [Accepted: 06/23/2010] [Indexed: 11/18/2022]
Abstract
Neospora caninum is an apicomplexan parasite, closely related to Toxoplasma gondii, and causes abortion and congenital neosporosis in cattle worldwide. Trophoblast cells act in mechanisms of innate immune defense at the fetal-maternal interface and no data are available about the interaction of Neospora with human trophoblasts. Thus, this study aimed to verify the susceptibility of human trophoblastic (BeWo) compared with uterine cervical (HeLa) cell lines to N. caninum. BeWo and HeLa cells were infected with different parasite:cell ratios of N. caninum tachyzoites and analyzed at different times after infection for cell viability using thiazolyl blue tetrazole and lactate dehydrogenase assays. Both cell lines were also evaluated for cytokine production and parasite infection/replication assays when pre-treated or not with Neospora lysate antigen (NLA) or human recombinant IFN-γ. Cell viability was increased up to 48 h of infection in both types of cells, suggesting that infection could inhibit early cell death and/or induce cell proliferation. Neospora infection induced up-regulation of the macrophage migration inhibitory factor (MIF), mainly in HeLa cells, which was enhanced by cell pre-treatment by NLA or IFN-γ. Conversely, parasite infection induced down-regulation of the transforming growth factor (TGF-β), mostly in BeWo cells, which was decreased with NLA or IFN-γ pre-treatment. HeLa cells were more susceptible to Neospora infection than BeWo cells and IFN-γ pre-treatment resulted in reduced infection indices in both cell lines. Control of parasite growth was mediated by IFN-γ through an indoleamine-2,3-dioxygenase-dependent mechanism in HeLa cells alone. Based on these results, we concluded that BeWo and HeLa cells are readily infected by N. caninum, although presenting differences in susceptibility to infection, cytokine production and cell viability. Thus, these host cells can be considered in comparative approaches to understand strategies used by N. caninum to survive at the maternal-fetal interface.
Collapse
Affiliation(s)
- Julianne V Carvalho
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Av. Pará 1720, 38400-902 Uberlândia, MG, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ortiz-Alegría LB, Caballero-Ortega H, Cañedo-Solares I, Rico-Torres CP, Sahagún-Ruiz A, Medina-Escutia ME, Correa D. Congenital toxoplasmosis: candidate host immune genes relevant for vertical transmission and pathogenesis. Genes Immun 2010; 11:363-73. [PMID: 20445562 DOI: 10.1038/gene.2010.21] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Toxoplasma gondii infects a variety of vertebrate hosts, including humans. Transplacental passage of the parasite leads to congenital toxoplasmosis. A primary infection during the first weeks of gestation causes vertical transmission at low rate, although it causes major damage to the embryo. Transmission frequency increases to near 80% by the end of pregnancy, but the proportion of ill newborns is low. For transmission and pathogenesis, the parasite genetics is certainly important. Several host innate and adaptative immune response genes are induced during infection in adults, which control the rapidly replicating tachyzoite. The T helper 1 (Th1) response is protective, although it has to be modulated to avoid inflammatory damage. Paradoxical observations on this response pattern in congenital toxoplasmosis have been reported, as it may be protective or deleterious, inducing sterile abortion or favoring parasite transplacental passage. Regarding pregnancy, an early Th1 microenvironment is important for control of infectious diseases and successful implantation, although it has to be regulated to support trophoblast survival. Polymorphism of genes involved in these parallel phenomena, such as Toll-like receptors (TLRs), adhesins, cytokines, chemokines or their receptors, immunoglobulins or Fc receptors (FcRs), might be important in susceptibility for T. gondii vertical transmission, abortion or fetal pathology. In this study some examples are presented and discussed.
Collapse
Affiliation(s)
- L B Ortiz-Alegría
- Laboratorio de Inmunología Experimental, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, SSA, México DF, Mexico
| | | | | | | | | | | | | |
Collapse
|
48
|
Flores M, Saavedra R, Bautista R, Viedma R, Tenorio EP, Leng L, Sánchez Y, Juárez I, Satoskar AA, Shenoy AS, Terrazas LI, Bucala R, Barbi J, Satoskar AR, Rodriguez-Sosa M. Macrophage migration inhibitory factor (MIF) is critical for the host resistance against Toxoplasma gondii. FASEB J 2008; 22:3661-71. [PMID: 18606868 DOI: 10.1096/fj.08-111666] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Macrophage migration inhibitory factor (MIF) exerts either a protective or a deleterious role in the immune response to different pathogens. We analyzed herein the role of MIF in the host control of toxoplasmosis using MIF(-/-) mice backcrossed to either the BALB/c or the C57BL/6 genetic backgrounds. Both, wild-type (WT) BALB/c and MIF(-/-) BALB/c mice were susceptible to infection with highly virulent RH as well as moderately virulent ME49 strains of T. gondii. MIF(-/-) mice, however, showed greater liver damage and more brain cysts, produced less proinflammatory cytokines, and succumbed significantly faster than WT mice. Bone marrow-derived dendritic cells (BMDCs) from MIF(-/-) mice produced less interleukin-1beta, interleukin-12, and tumor necrosis factor-alpha than WT BMDCs after stimulation with soluble Toxoplasma antigen (STAg). Similar observations were made in CD11c(+) low-density cells isolated from the spleens of MIF(-/-) mice challenged with STAg. MIF(-/-) C57BL/6 mice succumbed to ME49 infection faster than their WT counterparts. C57BL/6 mice that succumbed to infection with the ME49 strain produced less MIF than resistant BALB/c mice similarly infected. Interestingly, an analysis of brains from patients with cerebral toxoplasmosis showed low levels of MIF expression. Together, these findings demonstrate that MIF plays a critical role in mediating host resistance against T. gondii.
Collapse
Affiliation(s)
- Marcos Flores
- Unidad de Biomedicina, FES-Iztacala, UNAM. Av. de los Barrios #1, Los Reyes Iztacala, 54090 Tlalnepantla, Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|