Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 792
Filtrer
1.
Bioessays ; 46(7): e2400053, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38713161

RÉSUMÉ

Trypanosoma brucei is the causal agent of African Trypanosomiasis in humans and other animals. It maintains a long-term infection through an antigenic variation based population survival strategy. To proliferate in a mammal, T. brucei acquires iron and haem through the receptor mediated uptake of host transferrin and haptoglobin-hemoglobin respectively. The receptors are exposed to host antibodies but this does not lead to clearance of the infection. Here we discuss how the trypanosome avoids this fate in the context of recent findings on the structure and cell biology of the receptors.


Sujet(s)
Trypanosoma brucei brucei , Maladie du sommeil , Trypanosoma brucei brucei/immunologie , Trypanosoma brucei brucei/métabolisme , Humains , Animaux , Maladie du sommeil/immunologie , Maladie du sommeil/parasitologie , Haptoglobines/métabolisme , Récepteurs de surface cellulaire/métabolisme , Récepteurs de surface cellulaire/immunologie , Transferrine/métabolisme , Hémoglobines/métabolisme , Protéines de protozoaire/métabolisme , Protéines de protozoaire/immunologie , Interactions hôte-parasite/immunologie , Fer/métabolisme , Anticorps antiprotozoaires/immunologie
2.
Int Immunopharmacol ; 134: 112250, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38749335

RÉSUMÉ

Trypanosoma brucei, a causative agent of human and animal trypanosomiasis, regularly switches its major surface antigen to avoid elimination by the immune system. Toll-like receptor 9 (TLR9) is a key modulator for resistance to host-infective trypanosomes; however, the underlying molecular mechanism remains indistinct. Thus, we first approached the issue using Tlr9-mutant mice that render them non-responsive to TLR9 agonists. After infection, T cells in the spleens of Tlr9-mutant mice were analyzed by flow cytometry and a reduction in CD8+, CD4+ T, and NKT cells was observed in Tlr9-mutant mice compared to WT mice. We further found that the responses of inflammatory cytokines in the sera were reduced in Tlr9-mutant mice after T. brucei infection. The underlying molecular mechanism was that T. b. brucei DNA activated TLR9, which consequently upregulated the expression of p38 and ERK/MAPK, resulting in host resistance to trypanosome infection. In conclusion, these findings provide novel insights into the TLR9-mediated host responses to trypanosome infection.


Sujet(s)
Cytokines , Transduction du signal , Récepteur-9 de type Toll-like , Trypanosoma brucei brucei , Maladie du sommeil , Récepteur-9 de type Toll-like/métabolisme , Récepteur-9 de type Toll-like/agonistes , Animaux , Trypanosoma brucei brucei/immunologie , Maladie du sommeil/immunologie , Souris , Cytokines/métabolisme , Souris knockout , Souris de lignée C57BL , Humains
3.
Elife ; 122024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38655765

RÉSUMÉ

African trypanosomes replicate within infected mammals where they are exposed to the complement system. This system centres around complement C3, which is present in a soluble form in serum but becomes covalently deposited onto the surfaces of pathogens after proteolytic cleavage to C3b. Membrane-associated C3b triggers different complement-mediated effectors which promote pathogen clearance. To counter complement-mediated clearance, African trypanosomes have a cell surface receptor, ISG65, which binds to C3b and which decreases the rate of trypanosome clearance in an infection model. However, the mechanism by which ISG65 reduces C3b function has not been determined. We reveal through cryogenic electron microscopy that ISG65 has two distinct binding sites for C3b, only one of which is available in C3 and C3d. We show that ISG65 does not block the formation of C3b or the function of the C3 convertase which catalyses the surface deposition of C3b. However, we show that ISG65 forms a specific conjugate with C3b, perhaps acting as a decoy. ISG65 also occludes the binding sites for complement receptors 2 and 3, which may disrupt recruitment of immune cells, including B cells, phagocytes, and granulocytes. This suggests that ISG65 protects trypanosomes by combining multiple approaches to dampen the complement cascade.


Sujet(s)
Complément C3b , Complément C3b/métabolisme , Humains , Liaison aux protéines , Trypanosoma brucei brucei/immunologie , Trypanosoma brucei brucei/métabolisme , Protéines de protozoaire/métabolisme , Protéines de protozoaire/immunologie , Cryomicroscopie électronique , Sites de fixation , Complément C3/métabolisme , Complément C3/immunologie
4.
Cell Rep ; 37(5): 109923, 2021 11 02.
Article de Anglais | MEDLINE | ID: mdl-34731611

RÉSUMÉ

The dense variant surface glycoprotein (VSG) coat of African trypanosomes represents the primary host-pathogen interface. Antigenic variation prevents clearing of the pathogen by employing a large repertoire of antigenically distinct VSG genes, thus neutralizing the host's antibody response. To explore the epitope space of VSGs, we generate anti-VSG nanobodies and combine high-resolution structural analysis of VSG-nanobody complexes with binding assays on living cells, revealing that these camelid antibodies bind deeply inside the coat. One nanobody causes rapid loss of cellular motility, possibly due to blockage of VSG mobility on the coat, whose rapid endocytosis and exocytosis are mechanistically linked to Trypanosoma brucei propulsion and whose density is required for survival. Electron microscopy studies demonstrate that this loss of motility is accompanied by rapid formation and shedding of nanovesicles and nanotubes, suggesting that increased protein crowding on the dense membrane can be a driving force for membrane fission in living cells.


Sujet(s)
Membrane cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Anticorps à domaine unique/pharmacologie , Trypanocides/pharmacologie , Trypanosoma brucei brucei/effets des médicaments et des substances chimiques , Maladie du sommeil/traitement médicamenteux , Glycoprotéines de surface variables du trypanosome/immunologie , Animaux , Spécificité des anticorps , Sites de fixation des anticorps , Camélidés du Nouveau Monde/immunologie , Lignée cellulaire , Membrane cellulaire/immunologie , Membrane cellulaire/métabolisme , Membrane cellulaire/ultrastructure , Endocytose/effets des médicaments et des substances chimiques , Épitopes , Exocytose/effets des médicaments et des substances chimiques , Liaison aux protéines , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/métabolisme , Trypanocides/immunologie , Trypanocides/métabolisme , Trypanosoma brucei brucei/immunologie , Trypanosoma brucei brucei/métabolisme , Trypanosoma brucei brucei/ultrastructure , Maladie du sommeil/immunologie , Maladie du sommeil/métabolisme , Maladie du sommeil/parasitologie , Glycoprotéines de surface variables du trypanosome/métabolisme
5.
J Immunol ; 207(10): 2551-2560, 2021 11 15.
Article de Anglais | MEDLINE | ID: mdl-34635586

RÉSUMÉ

The protozoan parasite Trypanosoma brucei is the causative agent of the neglected tropical disease human African trypanosomiasis, otherwise known as sleeping sickness. Trypanosomes have evolved many immune-evasion mechanisms to facilitate their own survival, as well as prolonging host survival to ensure completion of the parasitic life cycle. A key feature of the bloodstream form of T. brucei is the secretion of aromatic keto acids, which are metabolized from tryptophan. In this study, we describe an immunomodulatory role for one of these keto acids, indole-3-pyruvate (I3P). We demonstrate that I3P inhibits the production of PGs in activated macrophages. We also show that, despite the reduction in downstream PGs, I3P augments the expression of cyclooxygenase (COX2). This increase in COX2 expression is mediated in part via inhibition of PGs relieving a negative-feedback loop on COX2. Activation of the aryl hydrocarbon receptor also participates in this effect. However, the increase in COX2 expression is of little functionality, as we also provide evidence to suggest that I3P targets COX activity. This study therefore details an evasion strategy by which a trypanosome-secreted metabolite potently inhibits macrophage-derived PGs, which might promote host and trypanosome survival.


Sujet(s)
Cyclooxygenase 2/métabolisme , Indoles/métabolisme , Macrophages/immunologie , Prostaglandines/métabolisme , Maladie du sommeil/immunologie , Animaux , Humains , Échappement immunitaire/immunologie , Indoles/immunologie , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Prostaglandines/immunologie , Trypanosoma brucei brucei/immunologie , Trypanosoma brucei brucei/métabolisme , Maladie du sommeil/métabolisme
6.
Microbiol Spectr ; 9(2): e0091521, 2021 10 31.
Article de Anglais | MEDLINE | ID: mdl-34704826

RÉSUMÉ

Trypanosoma brucei belongs to a genus of protists that cause life-threatening and economically important diseases of human and animal populations in Sub-Saharan Africa. T. brucei cells are covered in surface glycoproteins, some of which are used to escape the host immune system. Exo-/endocytotic trafficking of these and other molecules occurs via a single copy organelle called the flagellar pocket (FP). The FP is maintained and enclosed around the flagellum by the flagellar pocket collar (FPC). To date, the most important cytoskeletal component of the FPC is an essential calcium-binding, polymer-forming protein called TbBILBO1. In searching for novel tools to study this protein, we raised nanobodies (Nb) against purified, full-length TbBILBO1. Nanobodies were selected according to their binding properties to TbBILBO1, tested as immunofluorescence tools, and expressed as intrabodies (INb). One of them, Nb48, proved to be the most robust nanobody and intrabody. We further demonstrate that inducible, cytoplasmic expression of INb48 was lethal to these parasites, producing abnormal phenotypes resembling those of TbBILBO1 RNA interference (RNAi) knockdown. Our results validate the feasibility of generating functional single-domain antibody-derived intrabodies to target trypanosome cytoskeleton proteins. IMPORTANCE Trypanosoma brucei belongs to a group of important zoonotic parasites. We investigated how these organisms develop their cytoskeleton (the internal skeleton that controls cell shape) and focused on an essential protein (BILBO1) first described in T. brucei. To develop our analysis, we used purified BILBO1 protein to immunize an alpaca to make nanobodies (Nb). Nanobodies are derived from the antigen-binding portion of a novel antibody type found only in the camel and shark families of animals. Anti-BILBO1 nanobodies were obtained, and their encoding genes were inducibly expressed within the cytoplasm of T. brucei as intrabodies (INb). Importantly, INb48 expression rapidly killed parasites producing phenotypes normally observed after RNA knockdown, providing clear proof of principle. The importance of this study is derived from this novel approach, which can be used to study neglected and emerging pathogens as well as new model organisms, especially those that do not have the RNAi system.


Sujet(s)
Protéines de liaison au calcium/immunologie , Mort cellulaire/immunologie , Protéines du cytosquelette/immunologie , Anticorps à domaine unique/immunologie , Trypanosoma brucei brucei/immunologie , Protéines de liaison au calcium/antagonistes et inhibiteurs , Protéines de liaison au calcium/métabolisme , Flagelles/métabolisme , Interférence par ARN , Trypanosoma brucei brucei/métabolisme , Maladie du sommeil/parasitologie
7.
PLoS Pathog ; 17(10): e1009968, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34614031

RÉSUMÉ

Liver macrophages internalize circulating bloodborne parasites. It remains poorly understood how this process affects the fate of the macrophages and T cell responses in the liver. Here, we report that infection by Trypanosoma brucei induced depletion of macrophages in the liver, leading to the repopulation of CXCL16-secreting intrahepatic macrophages, associated with substantial accumulation of CXCR6+CD4+ T cells in the liver. Interestingly, disruption of CXCR6 signaling did not affect control of the parasitemia, but significantly enhanced the survival of infected mice, associated with reduced inflammation and liver injury. Infected CXCR6 deficient mice displayed a reduced accumulation of CD4+ T cells in the liver; adoptive transfer experiments suggested that the reduction of CD4+ T cells in the liver was attributed to a cell intrinsic property of CXCR6 deficient CD4+ T cells. Importantly, infected CXCR6 deficient mice receiving wild-type CD4+ T cells survived significantly shorter than those receiving CXCR6 deficient CD4+ T cells, demonstrating that CXCR6+CD4+ T cells promote the mortality. We conclude that infection of T. brucei leads to depletion and repopulation of liver macrophages, associated with a substantial influx of CXCR6+CD4+ T cells that mediates mortality.


Sujet(s)
Lymphocytes T CD4+/immunologie , Foie/immunologie , Macrophages/immunologie , Maladie du sommeil/immunologie , Animaux , Souris , Récepteurs CXCR6/immunologie , Trypanosoma brucei brucei/immunologie
8.
PLoS Negl Trop Dis ; 15(9): e0009814, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34587165

RÉSUMÉ

Anemia caused by trypanosome infection is poorly understood. Autoimmunity during Trypanosoma brucei infection was proposed to have a role during anemia, but the mechanisms involved during this pathology have not been elucidated. In mouse models and human patients infected with malaria parasites, atypical B-cells promote anemia through the secretion of autoimmune anti-phosphatidylserine (anti-PS) antibodies that bind to uninfected erythrocytes and facilitate their clearance. Using mouse models of two trypanosome infections, Trypanosoma brucei and Trypanosoma cruzi, we assessed levels of autoantibodies and anemia. Our results indicate that acute T. brucei infection, but not T. cruzi, leads to early increased levels of plasma autoantibodies against different auto antigens tested (PS, DNA and erythrocyte lysate) and expansion of atypical B cells (ABCs) that secrete these autoantibodies. In vitro studies confirmed that a lysate of T. brucei, but not T. cruzi, could directly promote the expansion of these ABCs. PS exposure on erythrocyte plasma membrane seems to be an important contributor to anemia by delaying erythrocyte recovery since treatment with an agent that prevents binding to it (Annexin V) ameliorated anemia in T. brucei-infected mice. Analysis of the plasma of patients with human African trypanosomiasis (HAT) revealed high levels of anti-PS antibodies that correlated with anemia. Altogether these results suggest a relation between autoimmunity against PS and anemia in both mice and patients infected with T. brucei.


Sujet(s)
Anémie/étiologie , Auto-immunité , Phosphatidylsérine/immunologie , Maladie du sommeil/immunologie , Adolescent , Adulte , Animaux , Autoanticorps/immunologie , Érythrocytes/immunologie , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Trypanosoma , Trypanosoma brucei brucei/immunologie , Maladie du sommeil/complications , Jeune adulte
9.
PLoS Pathog ; 17(9): e1009933, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34525131

RÉSUMÉ

Adipose tissue is one of the major reservoirs of Trypanosoma brucei parasites, the causative agent of sleeping sickness, a fatal disease in humans. In mice, the gonadal adipose tissue (AT) typically harbors 2-5 million parasites, while most solid organs show 10 to 100-fold fewer parasites. In this study, we tested whether the AT environment responds immunologically to the presence of the parasite. Transcriptome analysis of T. brucei infected adipose tissue revealed that most upregulated host genes are involved in inflammation and immune cell functions. Histochemistry and flow cytometry confirmed an increasingly higher number of infiltrated macrophages, neutrophils and CD4+ and CD8+ T lymphocytes upon infection. A large proportion of these lymphocytes effectively produce the type 1 effector cytokines, IFN-γ and TNF-α. Additionally, the adipose tissue showed accumulation of antigen-specific IgM and IgG antibodies as infection progressed. Mice lacking T and/or B cells (Rag2-/-, Jht-/-), or the signature cytokine (Ifng-/-) displayed a higher parasite load both in circulation and in the AT, demonstrating the key role of the adaptive immune system in both compartments. Interestingly, infections of C3-/- mice showed that while complement system is dispensable to control parasite load in the blood, it is necessary in the AT and other solid tissues. We conclude that T. brucei infection triggers a broad and robust immune response in the AT, which requires the complement system to locally reduce parasite burden.


Sujet(s)
Tissu adipeux/immunologie , Tissu adipeux/microbiologie , Trypanosoma brucei brucei/immunologie , Maladie du sommeil/immunologie , Animaux , Souris
10.
Sci Rep ; 11(1): 9856, 2021 05 10.
Article de Anglais | MEDLINE | ID: mdl-33972588

RÉSUMÉ

Infections with Trypanosoma brucei sp. are established after the injection of metacyclic trypomastigotes into the skin dermis by the tsetse fly vector. The parasites then gain access to the local lymphatic vessels to infect the local draining lymph nodes and disseminate systemically via the bloodstream. Macrophages are considered to play an important role in host protection during the early stage of systemic trypanosome infections. Macrophages are abundant in the skin dermis, but relatively little is known of their impact on susceptibility to intradermal (ID) trypanosome infections. We show that although dermal injection of colony stimulating factor 1 (CSF1) increased the local abundance of macrophages in the skin, this did not affect susceptibility to ID T. brucei infection. However, bacterial LPS-stimulation in the dermis prior to ID trypanosome infection significantly reduced disease susceptibility. In vitro assays showed that LPS-stimulated macrophage-like RAW264.7 cells had enhanced cytotoxicity towards T. brucei, implying that dermal LPS-treatment may similarly enhance the ability of dermal macrophages to eliminate ID injected T. brucei parasites in the skin. A thorough understanding of the factors that reduce susceptibility to ID injected T. brucei infections may lead to the development of novel strategies to help reduce the transmission of African trypanosomes.


Sujet(s)
Prédisposition aux maladies/immunologie , Lipopolysaccharides/immunologie , Macrophages/immunologie , Peau/immunologie , Maladie du sommeil/immunologie , Animaux , Modèles animaux de maladie humaine , Prédisposition aux maladies/microbiologie , Femelle , Humains , Injections intradermiques , Lipopolysaccharides/administration et posologie , Facteur de stimulation des colonies de macrophages/administration et posologie , Facteur de stimulation des colonies de macrophages/immunologie , Souris , Souris transgéniques , Cellules RAW 264.7 , Récepteur de facteur de croissance granulocyte-macrophage/génétique , Protéines recombinantes/administration et posologie , Protéines recombinantes/immunologie , Peau/microbiologie , Suidae , Trypanosoma brucei brucei/immunologie , Trypanosoma brucei brucei/pathogénicité , Maladie du sommeil/parasitologie
11.
mBio ; 12(1)2021 02 16.
Article de Anglais | MEDLINE | ID: mdl-33593983

RÉSUMÉ

Tumor necrosis factor (TNF)/inducible nitric oxide synthase (iNOS)-producing dendritic cells (Tip-DCs) have profound impacts on host immune responses during infections. The mechanisms regulating Tip-DC development remain largely unknown. Here, using a mouse model of infection with African trypanosomes, we show that a deficiency in interleukin-27 receptor (IL-27R) signaling results in escalated intrahepatic accumulation of Ly6C-positive (Ly6C+) monocytes and their differentiation into Tip-DCs. Blocking Tip-DC development significantly ameliorates liver injury and increases the survival of infected IL-27R-/- mice. Mechanistically, Ly6C+ monocyte differentiation into pathogenic Tip-DCs in infected IL-27R-/- mice is driven by a CD4+ T cell-interferon gamma (IFN-γ) axis via cell-intrinsic IFN-γ signaling. In parallel, hyperactive IFN-γ signaling induces cell death of Ly6C-negative (Ly6C-) monocytes in a cell-intrinsic manner, which in turn aggravates the development of pathogenic Tip-DCs due to the loss of the negative regulation of Ly6C- monocytes on Ly6C+ monocyte differentiation into Tip-DCs. Thus, IL-27 inhibits the dual-track exacerbation of Tip-DC development induced by a CD4+ T cell-IFN-γ axis. We conclude that IL-27 negatively regulates Tip-DC development by preventing the cell-intrinsic effects of IFN-γ and that the regulation involves CD4+ T cells and Ly6C- monocytes. Targeting IL-27 signaling may manipulate Tip-DC development for therapeutic intervention.IMPORTANCE TNF/iNOS-producing dendritic cells (Tip-DCs) are at the front line as immune effector cells to fight off a broad range of invading microbes. Excessive development of Tip-DCs contributes to tissue destruction. Thus, identifying master regulators of Tip-DC development is fundamental for developing new therapeutic strategies. Here, we identify Tip-DCs as a terminal target of IL-27, which prevents Tip-DC-mediated early mortality during parasitic infections. We demonstrate that IL-27 inhibits Tip-DC development via a dual-track mechanism involving the complex interactions of effector CD4+ T cells, Ly6C- monocytes, and Ly6C+ monocytes. These findings delineate an in-depth view of mechanisms of Tip-DC differentiation that may have significant implications for the ongoing development of IL-27-based immunotherapy.


Sujet(s)
Différenciation cellulaire/immunologie , Cellules dendritiques/physiologie , Régulation de l'expression des gènes , Interleukines/génétique , Nitric oxide synthase type II/immunologie , Récepteurs aux interleukines/génétique , Trypanosoma congolense/immunologie , Facteur de nécrose tumorale alpha/immunologie , Animaux , Lymphocytes T CD4+/immunologie , Cellules dendritiques/immunologie , Interféron gamma/génétique , Interféron gamma/immunologie , Interféron gamma/métabolisme , Interleukines/immunologie , Activation des lymphocytes , Souris , Souris de lignée C57BL , Souris knockout , Monocytes/immunologie , Monocytes/physiologie , Nitric oxide synthase type II/biosynthèse , Récepteurs aux interleukines/immunologie , Transduction du signal/immunologie , Trypanosoma brucei brucei/immunologie , Facteur de nécrose tumorale alpha/biosynthèse
12.
Mol Immunol ; 132: 172-183, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33601226

RÉSUMÉ

The trypanosomatid pathogens Leishmania spp., Trypanosoma cruzi, and Trypanosoma brucei, currently grouped as TriTryps, have evolved through the time to overcome the upfront innate immune response and establish the infection in humans adapting many aspects of the parasite-cell host interaction. Extracellular vesicles (EVs) emerge as critical structures carrying different key molecules from parasites and target cells that interact continuously during infection. Current information regarding the structure and composition of these vesicles provide new insights into the primary role of TriTryps-EVs reviewed in this work. Expanding knowledge about these critical vesicular structures will promote advances in basic sciences and in translational applications controlling pathogenesis in the neglected tropical diseases caused by TriTryps.


Sujet(s)
Vésicules extracellulaires/immunologie , Leishmania major/immunologie , Protozooses/immunologie , Trypanosoma brucei brucei/immunologie , Trypanosoma cruzi/immunologie , Animaux , Vésicules extracellulaires/parasitologie , Interactions hôte-parasite/immunologie , Humains , Immunité innée/immunologie , Protozooses/parasitologie
13.
Nucleic Acids Res ; 49(3): 1436-1454, 2021 02 22.
Article de Anglais | MEDLINE | ID: mdl-33450001

RÉSUMÉ

Homologous recombination dominates as the major form of DNA repair in Trypanosoma brucei, and is especially important for recombination of the subtelomeric variant surface glycoprotein during antigenic variation. RAD50, a component of the MRN complex (MRE11, RAD50, NBS1), is central to homologous recombination through facilitating resection and governing the DNA damage response. The function of RAD50 in trypanosomes is untested. Here we report that RAD50 and MRE11 are required for RAD51-dependent homologous recombination and phosphorylation of histone H2A following a DNA double strand break (DSB), but neither MRE11 nor RAD50 substantially influence DSB resection at a chromosome-internal locus. In addition, we reveal intrinsic separation-of-function between T. brucei RAD50 and MRE11, with only RAD50 suppressing DSB repair using donors with short stretches of homology at a subtelomeric locus, and only MRE11 directing DSB resection at the same locus. Finally, we show that loss of either MRE11 or RAD50 causes a greater diversity of expressed VSG variants following DSB repair. We conclude that MRN promotes stringent homologous recombination at subtelomeric loci and restrains antigenic variation.


Sujet(s)
Variation des antigènes , Protéines de liaison à l'ADN/physiologie , Protéine homologue de MRE11/physiologie , Protéines de protozoaire/physiologie , Réparation de l'ADN par recombinaison , Trypanosoma brucei brucei/génétique , Cassures double-brin de l'ADN , Trypanosoma brucei brucei/immunologie
14.
Sci China Life Sci ; 64(4): 621-632, 2021 Apr.
Article de Anglais | MEDLINE | ID: mdl-33420923

RÉSUMÉ

African trypanosomatid parasites escape host acquired immune responses through periodic antigenic variation of their surface coat. In this study, we describe a mechanism by which the parasites counteract innate immune responses. Two TatD DNases were identified in each of Trypanosoma evansi and Trypanosoma brucei. These DNases are bivalent metal-dependent endonucleases localized in the cytoplasm and flagella of the parasites that can also be secreted by the parasites. These enzymes possess conserved functional domains and have efficient DNA hydrolysis activity. Host neutrophil extracellular traps (NETs) induced by the parasites could be hydrolyzed by native and recombinant TatD DNases. NET disruption was prevented, and the survival rate of parasites was decreased, in the presence of the DNase inhibitor aurintricarboxylic acid. These data suggest that trypanosomes can counteract host innate immune responses by active secretion of TatD DNases to degrade NETs.


Sujet(s)
Désoxyribonucléases/immunologie , Pièges extracellulaires/immunologie , Échappement immunitaire/immunologie , Protéines de protozoaire/immunologie , Trypanosoma brucei brucei/immunologie , Trypanosoma/immunologie , Séquence d'acides aminés , Animaux , Désoxyribonucléases/génétique , Désoxyribonucléases/métabolisme , Pièges extracellulaires/métabolisme , Pièges extracellulaires/parasitologie , Femelle , Interactions hôte-pathogène/immunologie , Immunité innée/immunologie , Souris de lignée BALB C , Microscopie électronique à balayage , Microscopie immunoélectronique , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/parasitologie , Phylogenèse , Protozooses animales/immunologie , Protozooses animales/parasitologie , Protéines de protozoaire/classification , Protéines de protozoaire/métabolisme , Rat Sprague-Dawley , Similitude de séquences d'acides aminés , Spécificité d'espèce , Trypanosoma/métabolisme , Trypanosoma/ultrastructure , Trypanosoma brucei brucei/métabolisme , Trypanosoma brucei brucei/ultrastructure
15.
Nat Microbiol ; 6(3): 289-300, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33432154

RÉSUMÉ

Highly selective gene expression is a key requirement for antigenic variation in several pathogens, allowing evasion of host immune responses and maintenance of persistent infections1. African trypanosomes-parasites that cause lethal diseases in humans and livestock-employ an antigenic variation mechanism that involves monogenic antigen expression from a pool of >2,600 antigen-coding genes2. In other eukaryotes, the expression of individual genes can be enhanced by mechanisms involving the juxtaposition of otherwise distal chromosomal loci in the three-dimensional nuclear space3-5. However, trypanosomes lack classical enhancer sequences or regulated transcription initiation6,7. In this context, it has remained unclear how genome architecture contributes to monogenic transcription elongation and transcript processing. Here, we show that the single expressed antigen-coding gene displays a specific inter-chromosomal interaction with a major messenger RNA splicing locus. Chromosome conformation capture (Hi-C) revealed a dynamic reconfiguration of this inter-chromosomal interaction upon activation of another antigen. Super-resolution microscopy showed the interaction to be heritable and splicing dependent. We found a specific association of the two genomic loci with the antigen exclusion complex, whereby VSG exclusion 1 (VEX1) occupied the splicing locus and VEX2 occupied the antigen-coding locus. Following VEX2 depletion, loss of monogenic antigen expression was accompanied by increased interactions between previously silent antigen genes and the splicing locus. Our results reveal a mechanism to ensure monogenic expression, where antigen transcription and messenger RNA splicing occur in a specific nuclear compartment. These findings suggest a new means of post-transcriptional gene regulation.


Sujet(s)
Épissage des ARN/génétique , Transcription génétique/génétique , Trypanosoma brucei brucei/génétique , Glycoprotéines de surface variables du trypanosome/génétique , Variation des antigènes/génétique , Chromosomes/génétique , Chromosomes/métabolisme , Régulation de l'expression des gènes , Génome de protozoaire/génétique , Famille multigénique/génétique , ARN de tête épissé/génétique , Trypanosoma brucei brucei/immunologie
16.
Front Immunol ; 11: 559561, 2020.
Article de Anglais | MEDLINE | ID: mdl-33193328

RÉSUMÉ

Trypanosoma brucei brucei trypomastigotes are classical blood parasites of cattle, these stages might become potential targets for circulating polymorphonuclear neutrophils (PMN). We here investigated NETs extrusion and related oxygen consumption in bovine PMN exposed to motile T. b. brucei trypomastigotes in vitro. Parasite exposure induced PMN activation as detected by enhanced oxygen consumption rates (OCR), extracellular acidification rates (ECAR), and production of total and extracellular reactive oxygen species (ROS). Scanning electron microscopy (SEM) showed that co-cultivation of bovine PMN with motile trypomastigotes resulted in NETs formation within 120 min of exposure. T. b. brucei-induced NETs were confirmed by confocal microscopy demonstrating co-localization of extruded DNA with neutrophil elastase (NE) and nuclear histones. Immunofluorescence analyses demonstrated that trypomastigotes induced different phenotypes of NETs in bovine PMN, such as aggregated NETs (aggNETs), spread NETs (sprNETs), and diffuse NETs (diffNETs) with aggNETs being the most abundant ones. Furthermore, live cell 3D-holotomographic microscopy unveiled detailed morphological changes during the NETotic process. Quantification of T. b. brucei-induced NETs formation was estimated by DNA and nuclear area analysis (DANA) and confirmed enhanced NETs formation in response to trypomastigote stages. Formation of NETs does not result in a decrease of T. b. brucei viability, but a decrease of 26% in the number of motile parasites. Referring the involved signaling pathways, trypomastigote-induced NETs formation seems to be purinergic-dependent, since inhibition via NF449 treatment resulted in a significant reduction of T. b. brucei-triggered DNA extrusion. Overall, future studies will have to analyze whether the formation of aggNETs indeed plays a role in the outcome of clinical disease and bovine African trypanosomiasis-related immunopathological disorders, such as increased intravascular coagulopathy and vascular permeability, often reported to occur in this disease.


Sujet(s)
Maladies des bovins/immunologie , Maladies des bovins/parasitologie , Pièges extracellulaires/immunologie , Activation des neutrophiles/immunologie , Granulocytes neutrophiles/immunologie , Trypanosoma brucei brucei/immunologie , Maladie du sommeil/médecine vétérinaire , Animaux , Bovins , Pièges extracellulaires/métabolisme , Mitochondries/génétique , Mitochondries/métabolisme , NADPH oxidase/métabolisme , Granulocytes neutrophiles/métabolisme , Consommation d'oxygène , Espèces réactives de l'oxygène/métabolisme , Récepteurs purinergiques/métabolisme , Transduction du signal
17.
J Biol Chem ; 295(38): 13138-13149, 2020 09 18.
Article de Anglais | MEDLINE | ID: mdl-32727852

RÉSUMÉ

The human innate immunity factor apolipoprotein L-I (APOL1) protects against infection by several protozoan parasites, including Trypanosoma brucei brucei Endocytosis and acidification of high-density lipoprotein-associated APOL1 in trypanosome endosomes leads to eventual lysis of the parasite due to increased plasma membrane cation permeability, followed by colloid-osmotic swelling. It was previously shown that recombinant APOL1 inserts into planar lipid bilayers at acidic pH to form pH-gated nonselective cation channels that are opened upon pH neutralization. This corresponds to the pH changes encountered during endocytic recycling, suggesting APOL1 forms a cytotoxic cation channel in the parasite plasma membrane. Currently, the mechanism and domains required for channel formation have yet to be elucidated, although a predicted helix-loop-helix (H-L-H) was suggested to form pores by virtue of its similarity to bacterial pore-forming colicins. Here, we compare recombinant human and baboon APOL1 orthologs, along with interspecies chimeras and individual amino acid substitutions, to identify regions required for channel formation and pH gating in planar lipid bilayers. We found that whereas neutralization of glutamates within the H-L-H may be important for pH-dependent channel formation, there was no evidence of H-L-H involvement in either pH gating or ion selectivity. In contrast, we found two residues in the C-terminal domain, tyrosine 351 and glutamate 355, that influence pH gating properties, as well as a single residue, aspartate 348, that determines both cation selectivity and pH gating. These data point to the predicted transmembrane region closest to the APOL1 C terminus as the pore-lining segment of this novel channel-forming protein.


Sujet(s)
Apolipoprotéine L1/composition chimique , Immunité innée , Animaux , Apolipoprotéine L1/génétique , Apolipoprotéine L1/immunologie , Motifs à hélice-boucle-hélice , Humains , Concentration en ions d'hydrogène , Papio hamadryas , Trypanosoma brucei brucei/immunologie
18.
Methods Mol Biol ; 2116: 739-753, 2020.
Article de Anglais | MEDLINE | ID: mdl-32221952

RÉSUMÉ

Magnetic- and fluorescent-activated cell sorting (MACS and FACS) are used for isolation of distinct cell populations for subsequent studies including transcriptomics. The latter allows for the analysis of infection-induced alterations in gene expression profiles. MACS and FACS both use antibodies against cell surface molecules to isolate populations of interest. Standardized methods for both approaches exist for use in mouse models. These protocols, however, do not account for the fact that infection-associated immunopathology can significantly modulate the cell surface expression of targeted molecules. This is the case for Trypanosoma brucei brucei infection, where downregulation of CD23 surface expression on B cells has been reported. This hallmark of progressing infection interferes with the commercially available MACS technique for B cell purification, as CD23 expression is the target for the separation between Marginal Zone (MZ) and Follicular (Fo) B cells. Here, we provide a robust alternative method for isolation of infection-derived MZ B cells using CD1d and B220 surface molecules in a two-step MACS-FACS approach. The method yields 99% pure viable infection-derived MZ B cells, allowing extraction of a high quality total RNA suitable for subsequent RNA sequencing.


Sujet(s)
Lymphocytes B/métabolisme , Séparation cellulaire/méthodes , Trypanosoma brucei brucei/immunologie , Maladie du sommeil/diagnostic , Animaux , Antigène CD1d/génétique , Antigène CD1d/métabolisme , Lymphocytes B/immunologie , Séparation cellulaire/instrumentation , Modèles animaux de maladie humaine , Évolution de la maladie , Régulation négative/immunologie , Femelle , Cytométrie en flux/instrumentation , Cytométrie en flux/méthodes , Humains , Séparation immunomagnétique/instrumentation , Séparation immunomagnétique/méthodes , Antigènes CD45/génétique , Antigènes CD45/métabolisme , Souris , ARN/isolement et purification , RNA-Seq , Récepteurs aux IgE/génétique , Récepteurs aux IgE/métabolisme , Rate/cytologie , Rate/immunologie , Maladie du sommeil/immunologie , Maladie du sommeil/parasitologie
19.
Nat Commun ; 11(1): 844, 2020 02 12.
Article de Anglais | MEDLINE | ID: mdl-32051413

RÉSUMÉ

African trypanosomes (Trypanosoma) are vector-borne haemoparasites that survive in the vertebrate bloodstream through antigenic variation of their Variant Surface Glycoprotein (VSG). Recombination, or rather segmented gene conversion, is fundamental in Trypanosoma brucei for both VSG gene switching and for generating antigenic diversity during infections. Trypanosoma vivax is a related, livestock pathogen whose VSG lack structures that facilitate gene conversion in T. brucei and mechanisms underlying its antigenic diversity are poorly understood. Here we show that species-wide VSG repertoire is broadly conserved across diverse T. vivax clinical strains and has limited antigenic repertoire. We use variant antigen profiling, coalescent approaches and experimental infections to show that recombination plays little role in diversifying T. vivax VSG sequences. These results have immediate consequences for both the current mechanistic model of antigenic variation in African trypanosomes and species differences in virulence and transmission, requiring reconsideration of the wider epidemiology of animal African trypanosomiasis.


Sujet(s)
Variation des antigènes/génétique , Variation des antigènes/immunologie , Recombinaison génétique/génétique , Trypanosoma vivax/génétique , Glycoprotéines de surface variables du trypanosome/génétique , Glycoprotéines de surface variables du trypanosome/immunologie , ADN des protozoaires , Évolution moléculaire , Génome de protozoaire , Interactions hôte-parasite/immunologie , Échappement immunitaire , Phylogenèse , Protéines de protozoaire/génétique , Protéines de protozoaire/immunologie , Similitude de séquences , Spécificité d'espèce , Transcriptome , Trypanosoma brucei brucei/génétique , Trypanosoma brucei brucei/immunologie , Maladie du sommeil/immunologie , Maladie du sommeil/parasitologie , Glycoprotéines de surface variables du trypanosome/métabolisme
20.
Proc Natl Acad Sci U S A ; 117(5): 2613-2621, 2020 02 04.
Article de Anglais | MEDLINE | ID: mdl-31964820

RÉSUMÉ

Tsetse-transmitted African trypanosomes must develop into mammalian-infectious metacyclic cells in the fly's salivary glands (SGs) before transmission to a new host. The molecular mechanisms that underlie this developmental process, known as metacyclogenesis, are poorly understood. Blocking the few metacyclic parasites deposited in saliva from further development in the mammal could prevent disease. To obtain an in-depth perspective of metacyclogenesis, we performed single-cell RNA sequencing (scRNA-seq) from a pool of 2,045 parasites collected from infected tsetse SGs. Our data revealed three major cell clusters that represent the epimastigote, and pre- and mature metacyclic trypanosome developmental stages. Individual cell level data also confirm that the metacyclic pool is diverse, and that each parasite expresses only one of the unique metacyclic variant surface glycoprotein (mVSG) coat protein transcripts identified. Further clustering of cells revealed a dynamic transcriptomic and metabolic landscape reflective of a developmental program leading to infectious metacyclic forms preadapted to survive in the mammalian host environment. We describe the expression profile of proteins that regulate gene expression and that potentially play a role in metacyclogenesis. We also report on a family of nonvariant surface proteins (Fam10) and demonstrate surface localization of one member (named SGM1.7) on mature metacyclic parasites. Vaccination of mice with recombinant SGM1.7 reduced parasitemia early in the infection. Future studies are warranted to investigate Fam10 family proteins as potential trypanosome transmission blocking vaccine antigens. Our experimental approach is translationally relevant for developing strategies to prevent other insect saliva-transmitted parasites from infecting and causing disease in mammalian hosts.


Sujet(s)
Vecteurs insectes/parasitologie , Protéines de protozoaire/génétique , Trypanosoma brucei brucei/croissance et développement , Trypanosoma brucei brucei/génétique , Mouches tsé-tsé/parasitologie , Animaux , Femelle , Humains , Étapes du cycle de vie , Souris , Souris de lignée BALB C , Protéines de protozoaire/immunologie , ARN des protozoaires/génétique , Glandes salivaires/parasitologie , Analyse de séquence d'ARN , Analyse sur cellule unique , Transcriptome , Trypanosoma brucei brucei/immunologie , Maladie du sommeil/immunologie , Maladie du sommeil/parasitologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...