Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.020
Filtrer
1.
Int Immunopharmacol ; 140: 112847, 2024 Oct 25.
Article de Anglais | MEDLINE | ID: mdl-39088922

RÉSUMÉ

Trypanosomes are the extracellular protozoan parasites that cause human African trypanosomiasis disease in humans and nagana disease in animals. Tsetse flies act as a vector for the transmission of the disease in African countries. Animals infected with these parasites become useless or workless, and if not treated, disease can be fatal. There are many side effects associated with old treatments and some of them result in death in 5% of cases. There is a major surface glycoprotein in the parasite known as variant surface glycoprotein. The immune system of the host develops antibodies against this antigen but due to antigenic variation, parasites evade the immune response. Currently, no vaccine is available that provides complete protection. In murine models, only partial protection was observed using certain antigens. In order to develop vaccines against trypanosomes, molecular biology and immunology tools have been used. Immunization is the sole method for the control of disease because the eradication of the vector from endemic areas is an impossible task. Genetic vaccines can carry multiple genes encoding different antigens of the same parasite or different parasites. DNA immunization induces the activation of both cellular immune response and humoral immune response along with the generation of memory. This review highlights the importance of DNA vaccines and advances in the development of DNA vaccines against T. brucei.


Sujet(s)
Vaccins antiprotozoaires , Trypanosoma brucei brucei , Maladie du sommeil , Vaccins à ADN , Animaux , Vaccins à ADN/immunologie , Humains , Maladie du sommeil/prévention et contrôle , Maladie du sommeil/immunologie , Trypanosoma brucei brucei/immunologie , Trypanosoma brucei brucei/génétique , Vaccins antiprotozoaires/immunologie , Développement de vaccin , Antigènes de protozoaire/immunologie
2.
Front Immunol ; 15: 1441131, 2024.
Article de Anglais | MEDLINE | ID: mdl-39114668

RÉSUMÉ

Haptoglobin is a plasma protein of mammals that plays a crucial role in vascular homeostasis by binding free haemoglobin released from ruptured red blood cells. Trypanosoma brucei can exploit this by internalising haptoglobin-haemoglobin complex to acquire host haem. Here, we investigated the impact of haptoglobin deficiency (Hp-/-) on T. brucei brucei infection and the parasite´s capacity to internalise haemoglobin in a Hp-/- mouse model. The infected Hp-/- mice exhibited normal disease progression, with minimal weight loss and no apparent organ pathology, similarly to control mice. While the proteomic profile of mouse sera significantly changed in response to T. b. brucei, no differences in the infection response markers of blood plasma between Hp-/- and control Black mice were observed. Similarly, very few quantitative differences were observed between the proteomes of parasites harvested from Hp-/- and Black mice, including both endogenous proteins and internalised host proteins. While haptoglobin was indeed absent from parasites isolated from Hp-/-mice, haemoglobin peptides were unexpectedly detected in parasites from both Hp-/- and Black mice. Combined, the data support the dispensability of haptoglobin for haemoglobin internalisation by T. b. brucei during infection in mice. Since the trypanosomes knock-outs for their haptoglobin-haemoglobin receptor (HpHbR) internalised significantly less haemoglobin from Hp-/- mice compared to those isolated from Black mice, it suggests that T. b. brucei employs also an HpHbR-independent haptoglobin-mediated mode for haemoglobin internalisation. Our study reveals a so-far hidden flexibility of haemoglobin acquisition by T. b. brucei and offers novel insights into alternative haemoglobin uptake pathways.


Sujet(s)
Haptoglobines , Hémoglobines , Souris knockout , Trypanosoma brucei brucei , Maladie du sommeil , Animaux , Souris , Modèles animaux de maladie humaine , Haptoglobines/génétique , Haptoglobines/métabolisme , Hémoglobines/métabolisme , Souris de lignée C57BL , Protéomique/méthodes , Trypanosoma brucei brucei/métabolisme , Maladie du sommeil/parasitologie , Maladie du sommeil/immunologie , Mâle , Femelle
3.
PLoS Negl Trop Dis ; 18(7): e0012294, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38950072

RÉSUMÉ

The scarcity of reliable devices for diagnosis of Animal African trypanosomiasis (AAT) presents a limitation to control of the disease. Existing high-sensitivity technologies such as PCR are costly, laborious, time-consuming, complex, and require skilled personnel. Hence, utilisation of most diagnostics for AAT is impracticable in rural areas, where the disease occurs. A more accessible point-of-care test (POCT) capable of detecting cryptic active infection, without relying on expensive equipment, would facilitate AAT detection. In turn, early management, would reduce disease incidence and severity. Today, several ongoing research projects aim at modifying complex immunoassays into POCTs. In this context, we report the development of an antigen (Ag) detection sandwich ELISA prototype for diagnosis of T. congolense infections, which is comprised of nanobody (Nb) and monoclonal antibody (mAb) reagents. The Nb474H used here, originated from a past study. Briefly, the Nb was engineered starting from mRNA of peripheral blood lymphocytes of an alpaca immunized with soluble lysate of Trypanosoma congolense (TC13). T. congolense glycosomal fructose-1,6-bisphosphate aldolase (TcoALD) was discovered as the cognate Ag of Nb474H. In this study, splenocytes were harvested from a mouse immunized with recombinant TcoALD and fused with NS01 cells to generate a hybridoma library. Random screening of the library on TcoALD retrieved a lone binder, designated IgM8A2. Using Nb474H as Ag-capture reagent in combination with the IgM8A2 monoclonal antibody Ag-detection reagent resulted in a tool that effectively detects native TcoALD released during infection by T. congolense parasites. Hitherto, development of POCT for detection of active trypanosome infection is elusive. The Nanobody/Monoclonal Antibody (Nb/mAb) "hybrid" sandwich technology offers prospects for exploration, using the unique specificity of Nb as a key determinant in Ag capturing, while using the versatility of monoclonal Ab to adapt to various detection conditions.


Sujet(s)
Anticorps monoclonaux , Anticorps antiprotozoaires , Test ELISA , Trypanosoma congolense , Maladie du sommeil , Maladie du sommeil/diagnostic , Maladie du sommeil/immunologie , Animaux , Trypanosoma congolense/immunologie , Anticorps monoclonaux/immunologie , Anticorps antiprotozoaires/sang , Anticorps antiprotozoaires/immunologie , Test ELISA/méthodes , Souris , Anticorps à domaine unique/immunologie , Antigènes de protozoaire/immunologie , Sensibilité et spécificité
4.
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
5.
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
6.
PLoS Pathog ; 20(4): e1012186, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38648216

RÉSUMÉ

In the bloodstream of mammalian hosts, African trypanosomes face the challenge of protecting their invariant surface receptors from immune detection. This crucial role is fulfilled by a dense, glycosylated protein layer composed of variant surface glycoproteins (VSGs), which undergo antigenic variation and provide a physical barrier that shields the underlying invariant surface glycoproteins (ISGs). The protective shield's limited permeability comes at the cost of restricted access to the extracellular host environment, raising questions regarding the specific function of the ISG repertoire. In this study, we employ an integrative structural biology approach to show that intrinsically disordered membrane-proximal regions are a common feature of members of the ISG super-family, conferring the ability to switch between compact and elongated conformers. While the folded, membrane-distal ectodomain is buried within the VSG layer for compact conformers, their elongated counterparts would enable the extension beyond it. This dynamic behavior enables ISGs to maintain a low immunogenic footprint while still allowing them to engage with the host environment when necessary. Our findings add further evidence to a dynamic molecular organization of trypanosome surface antigens wherein intrinsic disorder underpins the characteristics of a highly flexible ISG proteome to circumvent the constraints imposed by the VSG coat.


Sujet(s)
Maladie du sommeil , Glycoprotéines de surface variables du trypanosome , Glycoprotéines de surface variables du trypanosome/métabolisme , Maladie du sommeil/parasitologie , Maladie du sommeil/immunologie , Protéines de protozoaire/métabolisme , Humains , Glycoprotéines membranaires/métabolisme , Animaux
7.
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
8.
PLoS Negl Trop Dis ; 15(11): e0009892, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34762691

RÉSUMÉ

Human African trypanosomiasis (HAT), also known as sleeping sickness, is a major cause of mortality and morbidity in sub-Saharan Africa. We hypothesised that recent findings of neurological features and parasite brain infiltration occurring at much earlier stages in HAT than previously thought could be explained by early activation of host genetic programmes controlling CNS disease. Accordingly, a transcriptomal analysis was performed on brain tissue at 0, 7, 14, 21 and 28dpi from the HAT CD1/GVR35 mouse model. Up to 21dpi, most parasites are restricted to the blood and lymphatic system. Thereafter the trypanosomes enter the brain initiating the encephalitic stage. Analysis of ten different time point Comparison pairings, revealed a dynamic transcriptome comprising four message populations. All 7dpi Comparisons had by far more differentially expressed genes compared to all others. Prior to invasion of the parenchyma, by 7dpi, ~2,000 genes were up-regulated, denoted [7dpi↑] in contrast to a down regulated population [7dpi↓] also numbering ~2,000. However, by 14dpi both patterns had returned to around the pre-infected levels. The third, [28dpi↑] featured over three hundred transcripts which had increased modestly up to14dpi, thereafter were significantly up-regulated and peaked at 28dpi. The fourth, a minor population, [7dpi↑-28dpi↑], had similar elevated levels at 7dpi and 28dpi. KEGG and GO enrichment analysis predicted a diverse phenotype by 7dpi with changes to innate and adaptive immunity, a Type I interferon response, neurotransmission, synaptic plasticity, pleiotropic signalling, circadian activity and vascular permeability without disruption of the blood brain barrier. This key observation is consistent with recent rodent model neuroinvasion studies and clinical reports of Stage 1 HAT patients exhibiting CNS symptoms. Together, these findings challenge the strict Stage1/Stage2 phenotypic demarcation in HAT and show that that significant neurological, and immune changes can be detected prior to the onset of CNS disease.


Sujet(s)
Encéphale/parasitologie , Maladies du système nerveux central/génétique , Maladies du système nerveux central/immunologie , Trypanosoma brucei brucei/physiologie , Maladie du sommeil/génétique , Maladie du sommeil/immunologie , Animaux , Barrière hémato-encéphalique/immunologie , Barrière hémato-encéphalique/parasitologie , Encéphale/immunologie , Maladies du système nerveux central/parasitologie , Modèles animaux de maladie humaine , Évolution de la maladie , Femelle , Analyse de profil d'expression de gènes , Humains , Souris , Phénotype , Analyse par réseau de protéines , Transcription génétique , Maladie du sommeil/parasitologie
9.
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
10.
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
11.
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
12.
PLoS Negl Trop Dis ; 15(9): e0009764, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34587172

RÉSUMÉ

The infection by Trypanosoma brucei brucei (T.b.b.), a protozoan parasite, is characterized by an early-systemic stage followed by a late stage in which parasites invade the brain parenchyma in a T cell-dependent manner. Here we found that early after infection effector-memory T cells were predominant among brain T cells, whereas, during the encephalitic stage T cells acquired a tissue resident memory phenotype (TRM) and expressed PD1. Both CD4 and CD8 T cells were independently redundant for the penetration of T.b.b. and other leukocytes into the brain parenchyma. The role of lymphoid cells during the T.b.b. infection was studied by comparing T- and B-cell deficient rag1-/- and WT mice. Early after infection, parasites located in circumventricular organs, brain structures with increased vascular permeability, particularly in the median eminence (ME), paced closed to the sleep-wake regulatory arcuate nucleus of the hypothalamus (Arc). Whereas parasite levels in the ME were higher in rag1-/- than in WT mice, leukocytes were instead reduced. Rag1-/- infected mice showed increased levels of meca32 mRNA coding for a blood /hypothalamus endothelial molecule absent in the blood-brain-barrier (BBB). Both immune and metabolic transcripts were elevated in the ME/Arc of WT and rag1-/- mice early after infection, except for ifng mRNA, which levels were only increased in WT mice. Finally, using a non-invasive sleep-wake cycle assessment method we proposed a putative role of lymphocytes in mediating sleep alterations during the infection with T.b.b. Thus, the majority of T cells in the brain during the early stage of T.b.b. infection expressed an effector-memory phenotype while TRM cells developed in the late stage of infection. T cells and parasites invade the ME/Arc altering the metabolic and inflammatory responses during the early stage of infection and modulating sleep disturbances.


Sujet(s)
Maladies du système nerveux central/immunologie , Maladies du système nerveux central/parasitologie , Sous-populations de lymphocytes T/physiologie , Trypanosoma brucei brucei , Maladie du sommeil/immunologie , Maladie du sommeil/anatomopathologie , Animaux , Encéphale/parasitologie , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Humains , Mémoire immunologique , Leucocytes , Souris , Souris knockout , Sommeil
13.
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
14.
J Immunol ; 207(5): 1401-1410, 2021 09 01.
Article de Anglais | MEDLINE | ID: mdl-34380646

RÉSUMÉ

PI3Kδ is critical in generating humoral and regulatory immune responses. In this study, we determined the impact of PI3Kδ in immunity to Trypanosoma congolense, an African trypanosome that can manipulate and evade Ab responses critical for protection. Upon infection with T. congolense, PI3KδD910A mice lacking PI3Kδ activity paradoxically show a transient enhancement in early control of parasitemia, associated with impaired production of regulatory IL-10 by B cells in the peritoneum. C57BL/6 wild-type (WT) mice treated with the PI3Kδ inhibitor (PI3Kδi) Idelalisib showed a similar transient decrease in parasitemia associated with reduced IL-10. Strikingly, however, we find that PI3KδD910A mice were ultimately unable to control this infection, resulting in uncontrolled parasitemia and death within 2 wk. Assessment of humoral responses revealed delayed B cell activation, impaired germinal center responses, and compromised Ab responses to differing degrees in PI3KδD910A and PI3Kδi-treated mice. To test the role of Abs, we administered serum from WT mice to PI3KδD910A mice and found that lethality was prevented by postinfection serum. Interestingly, serum from naive WT mice provided partial protection to PI3KδD910A mutants, indicating an additional role for natural Abs. Together our findings suggest that although PI3Kδ drives immune regulatory responses that antagonize early control of parasite growth in the peritoneum, it is also required for generation of Abs that are critical for protection from systemic trypanosome infection. The essential role of PI3Kδ for host survival of African trypanosome infection contrasts with findings for other pathogens such as Leishmania, underlining the critical importance of PI3Kδ-dependent humoral immunity in this disease.


Sujet(s)
Lymphocytes B/immunologie , Phosphatidylinositol 3-kinases de classe I/métabolisme , Trypanosoma congolense/physiologie , Maladie du sommeil/immunologie , Animaux , Phosphatidylinositol 3-kinases de classe I/génétique , Immunité humorale , Immunomodulation , Interleukine-10/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Parasitémie
15.
Am J Trop Med Hyg ; 105(5): 1193-1197, 2021 08 09.
Article de Anglais | MEDLINE | ID: mdl-34370709

RÉSUMÉ

The recent introduction of large-scale, population-based serologic surveys in several nations where human African trypanosomiasis (HAT) remains endemic could provide an opportunity to better map the remaining disease foci and to identify asymptomatic, seropositive individuals who are infected with the more chronic form of the parasite, Trypanosoma brucei gambiense (gHAT). We have incorporated a soluble form of variant surface glycoprotein 117 and a recombinant invariant surface glycoprotein 65.1 into a multiplex bead assay (MBA) method that is commonly used for the detection of IgG antibody responses to other neglected tropical diseases. A positive result was defined as reactivity to both antigens. MBA sensitivity and specificity for gHAT infection were 92% and 96%, respectively. Assay specificity for the acute form of disease caused by T.b. rhodesiense (rHAT) was 94%, but the sensitivity was only 63.6%. In the future, additional antigens could be incorporated into the multiplex assay to improve rHAT sensitivity.


Sujet(s)
Production d'anticorps , Antigènes de protozoaire/sang , Antigènes de protozoaire/immunologie , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Trypanosoma brucei gambiense/immunologie , Maladie du sommeil/sang , Maladie du sommeil/immunologie , Humains , Sensibilité et spécificité , Maladie du sommeil/épidémiologie
16.
PLoS Negl Trop Dis ; 15(6): e0009526, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-34153047

RÉSUMÉ

Trypanosoma brucei rhodesiense is one of the causative agents of Human African Trypanosomiasis (HAT), known as sleeping sickness. The parasite invades the central nervous system and causes severe encephalitis that is fatal if left untreated. We have previously identified ecotin-like inhibitors of serine peptidases, named ISPs, in trypanosomatid parasitic protozoa. Here, we investigated the role of ISP2 in bloodstream form T. b. rhodesiense. We generated gene-deficient mutants lacking ISP2 (Δisp2), which displayed a growth profile in vitro similar to that of wild-type (WT) parasites. C57BL/6 mice infected with Δisp2 displayed lower blood parasitemia, a delayed hind leg pathological phenotype and survived longer. The immune response was examined at two time-points that corresponded with two peaks of parasitemia. At 4 days, the spleens of Δisp2-infected mice had a greater percentage of NOS2+ myeloid cells, IFN-γ+-NK cells and increased TNF-α compared to those infected with WT and parasites re-expressing ISP2 (Δisp2:ISP2). By 13 days the increased NOS2+ population was sustained in Δisp2-infected mice, along with increased percentages of monocyte-derived dendritic cells, as well as CD19+ B lymphocytes, and CD8+ and CD4+ T lymphocytes. Taken together, these findings indicate that ISP2 contributes to T. b. rhodesiense virulence in mice and attenuates the inflammatory response during early infection.


Sujet(s)
Inhibiteurs de la sérine protéinase/métabolisme , Trypanosoma brucei rhodesiense/génétique , Trypanosoma brucei rhodesiense/pathogénicité , Maladie du sommeil/immunologie , Animaux , Animal génétiquement modifié , Anticorps monoclonaux , Femelle , Inflammation , Souris de lignée C57BL , Inhibiteurs de la sérine protéinase/génétique , Rate/parasitologie , Virulence
17.
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
18.
Nature ; 595(7865): 96-100, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-34040257

RÉSUMÉ

Trypanosomes are protozoan parasites that cause infectious diseases, including African trypanosomiasis (sleeping sickness) in humans and nagana in economically important livestock1,2. An effective vaccine against trypanosomes would be an important control tool, but the parasite has evolved sophisticated immunoprotective mechanisms-including antigenic variation3-that present an apparently insurmountable barrier to vaccination. Here we show, using a systematic genome-led vaccinology approach and a mouse model of Trypanosoma vivax infection4, that protective invariant subunit vaccine antigens can be identified. Vaccination with a single recombinant protein comprising the extracellular region of a conserved cell-surface protein that is localized to the flagellum membrane (which we term 'invariant flagellum antigen from T. vivax') induced long-lasting protection. Immunity was passively transferred with immune serum, and recombinant monoclonal antibodies to this protein could induce sterile protection and revealed several mechanisms of antibody-mediated immunity, including a major role for complement. Our discovery identifies a vaccine candidate for an important parasitic disease that has constrained socioeconomic development in countries in sub-Saharan Africa5, and provides evidence that highly protective vaccines against trypanosome infections can be achieved.


Sujet(s)
Antigènes de protozoaire/immunologie , Vaccins antiprotozoaires/immunologie , Trypanosoma vivax/immunologie , Maladie du sommeil/immunologie , Maladie du sommeil/prévention et contrôle , Animaux , Antigènes de protozoaire/composition chimique , Protéines du système du complément/immunologie , Séquence conservée/immunologie , Modèles animaux de maladie humaine , Femelle , Flagelles/composition chimique , Flagelles/immunologie , Souris , Souris de lignée BALB C , Vaccins antiprotozoaires/composition chimique , Facteurs temps , Trypanosoma vivax/composition chimique , Trypanosoma vivax/cytologie , Maladie du sommeil/parasitologie , Vaccins sous-unitaires/composition chimique , Vaccins sous-unitaires/immunologie
19.
Infect Genet Evol ; 87: 104636, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33217546

RÉSUMÉ

Fighting trypanosomiasis with an anti-trypanosome vaccine is ineffective, the parasite being protected by a Variable Surface Glycoprotein (VSG) whose structure is modified at each peak of parasitaemia, which allows it to escape the host's immune defenses. However, the host immunization against an essential factor for the survival of the parasite or the expression of its pathogenicity could achieve the same objective. Here we present the results of mouse immunization against the Translationally Controlled Tumor Protein (TCTP), a protein present in the Trypanosoma brucei gambiense (Tbg) secretome, the parasite responsible for human trypanosomiasis. Mice immunization was followed by infection with Tbg parasites. The production of IgG, IgG1 and IgG2a begun after the second TCTP injection and was dose-dependant, the maximum level of anti-TCTP antibodies remained stable up to 4 days post-infection and then decreased. Regarding cytokines (IL-2, 4, 6, 10, INFγ, TNFα), the most striking result was their total suppression after immunization with the highest TCTP dose. Compared to the control group, the immunized mice displayed a reduced first peak of parasitaemia, a 100% increase in the time to onset of the second peak, and an increased time of mice survival. The effect of immunization was only transient but demonstrated the likely important role that TCTP plays in host-parasite interactions and that some key parasite proteins could reduce infection impact.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Cytokines/biosynthèse , Immunoglobulines/biosynthèse , Souris/parasitologie , Trypanosoma brucei gambiense/génétique , Trypanosoma brucei gambiense/pathogénicité , Maladie du sommeil/immunologie , Animaux , Cytokines/génétique , Modèles animaux de maladie humaine , Expression des gènes , Humains , Immunoglobulines/génétique , Protéine tumorale-1 contrôlée par la traduction
20.
Front Immunol ; 11: 1085, 2020.
Article de Anglais | MEDLINE | ID: mdl-32655552

RÉSUMÉ

In many infectious diseases, the immune response operates as a double-edged sword. While required for protective immunity, infection-induced inflammation can be detrimental if it is not properly controlled, causing collateral body damage and potentially leading to death. It is in this context that the potent anti-inflammatory cytokine interleukin-10 (IL-10) is required to dampen the pro-inflammatory immune response that hallmarks trypanosomosis. Effective control of this infection requires not just the action of antibodies specific for the parasite's variable surface glycoprotein (VSG) coat antigens, but also a pro-inflammatory immune response mediated mainly by IFNγ, TNF, and NO. However, strict control of inflammation is mandatory, as IL-10-deficient mice succumb from an unrestrained cytokine storm within 10 days of a Trypanosome brucei infection. The relevant cellular source of IL-10 and the associated molecular mechanisms implicated in its trypanosomosis associated production are poorly understood. Using an IL-10 reporter mouse strain (Vert-X), we demonstrate here that NK cells, CD8+ T cells and CD4+ T cells as well as B cells and plasma cells constitute potential cellular sources of IL-10 within the spleen and liver during acute infection. The IL-10 wave follows peak pro-inflammatory cytokine production, which accompanied the control of peak parasitemia. Similar results were observed following conventional experimental needle infection and physiological infections via T. brucei-infected tsetse flies. Our results show that conditional T cell-specific ablation of the IL-10 regulating Prdm1 gene (encoding for the Blimp-1 transcription factor), leads to an uncontrolled trypanosome-induced pro-inflammatory syndrome like the one observed in infected IL-10-deficient mice. This result indicates that the biological role of IL-10-derived from non-T cells, including NK cells, is of minor importance when considering host survival. The cytokine IL-27 that is also considered to be an IL-10 regulator, did not affect IL-10 production during infection. Together, these data suggest that T. brucei activates a Blimp-1-dependent IL-10 regulatory pathway in T cells that acts as a critical anti-inflammatory rheostat, mandatory for host survival during the acute phase of parasitemia.


Sujet(s)
Syndrome de libération de cytokines/prévention et contrôle , Interleukine-10/biosynthèse , Facteur-1 liant le domaine de régulation positive I/immunologie , Lymphocytes T/immunologie , Trypanosoma brucei brucei , Maladie du sommeil/immunologie , Animaux , Syndrome de libération de cytokines/étiologie , Syndrome de libération de cytokines/immunologie , Modèles animaux de maladie humaine , Femelle , Inflammation/étiologie , Inflammation/immunologie , Inflammation/prévention et contrôle , Vecteurs insectes/parasitologie , Interleukine-10/déficit , Interleukine-10/génétique , Interleukines/antagonistes et inhibiteurs , Interleukines/déficit , Interleukines/immunologie , Foie/immunologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Facteur-1 liant le domaine de régulation positive I/déficit , Facteur-1 liant le domaine de régulation positive I/génétique , Rate/immunologie , Maladie du sommeil/complications , Maladie du sommeil/parasitologie , Mouches tsé-tsé/parasitologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE