Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 790
Filtrar
1.
Elife ; 122024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38655765

RESUMO

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.


Assuntos
Complemento C3b , Complemento C3b/metabolismo , Humanos , Ligação Proteica , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei brucei/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/imunologia , Microscopia Crioeletrônica , Sítios de Ligação , Complemento C3/metabolismo , Complemento C3/imunologia
2.
Cell Rep ; 37(5): 109923, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731611

RESUMO

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.


Assuntos
Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Anticorpos de Domínio Único/farmacologia , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Tripanossomíase Africana/tratamento farmacológico , Glicoproteínas Variantes de Superfície de Trypanosoma/imunologia , Animais , Especificidade de Anticorpos , Sítios de Ligação de Anticorpos , Camelídeos Americanos/imunologia , Linhagem Celular , Membrana Celular/imunologia , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Endocitose/efeitos dos fármacos , Epitopos , Exocitose/efeitos dos fármacos , Ligação Proteica , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/metabolismo , Tripanossomicidas/imunologia , Tripanossomicidas/metabolismo , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/ultraestrutura , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/metabolismo , Tripanossomíase Africana/parasitologia , Glicoproteínas Variantes de Superfície de Trypanosoma/metabolismo
3.
PLoS Pathog ; 17(10): e1009968, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34614031

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fígado/imunologia , Macrófagos/imunologia , Tripanossomíase Africana/imunologia , Animais , Camundongos , Receptores CXCR6/imunologia , Trypanosoma brucei brucei/imunologia
4.
J Immunol ; 207(10): 2551-2560, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34635586

RESUMO

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.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Indóis/metabolismo , Macrófagos/imunologia , Prostaglandinas/metabolismo , Tripanossomíase Africana/imunologia , Animais , Humanos , Evasão da Resposta Imune/imunologia , Indóis/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Prostaglandinas/imunologia , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei brucei/metabolismo , Tripanossomíase Africana/metabolismo
5.
Microbiol Spectr ; 9(2): e0091521, 2021 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-34704826

RESUMO

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.


Assuntos
Proteínas de Ligação ao Cálcio/imunologia , Morte Celular/imunologia , Proteínas do Citoesqueleto/imunologia , Anticorpos de Domínio Único/imunologia , Trypanosoma brucei brucei/imunologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/metabolismo , Flagelos/metabolismo , Interferência de RNA , Trypanosoma brucei brucei/metabolismo , Tripanossomíase Africana/parasitologia
6.
PLoS Pathog ; 17(9): e1009933, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34525131

RESUMO

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.


Assuntos
Tecido Adiposo/imunologia , Tecido Adiposo/microbiologia , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/imunologia , Animais , Camundongos
7.
PLoS Negl Trop Dis ; 15(9): e0009814, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34587165

RESUMO

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.


Assuntos
Anemia/etiologia , Autoimunidade , Fosfatidilserinas/imunologia , Tripanossomíase Africana/imunologia , Adolescente , Adulto , Animais , Autoanticorpos/imunologia , Eritrócitos/imunologia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Trypanosoma , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/complicações , Adulto Jovem
8.
Sci Rep ; 11(1): 9856, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33972588

RESUMO

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.


Assuntos
Suscetibilidade a Doenças/imunologia , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Pele/imunologia , Tripanossomíase Africana/imunologia , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças/microbiologia , Feminino , Humanos , Injeções Intradérmicas , Lipopolissacarídeos/administração & dosagem , Fator Estimulador de Colônias de Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Células RAW 264.7 , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Pele/microbiologia , Suínos , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei brucei/patogenicidade , Tripanossomíase Africana/parasitologia
9.
mBio ; 12(1)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33593983

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/fisiologia , Regulação da Expressão Gênica , Interleucinas/genética , Óxido Nítrico Sintase Tipo II/imunologia , Receptores de Interleucina/genética , Trypanosoma congolense/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucinas/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Monócitos/fisiologia , Óxido Nítrico Sintase Tipo II/biossíntese , Receptores de Interleucina/imunologia , Transdução de Sinais/imunologia , Trypanosoma brucei brucei/imunologia , Fator de Necrose Tumoral alfa/biossíntese
10.
Mol Immunol ; 132: 172-183, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33601226

RESUMO

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.


Assuntos
Vesículas Extracelulares/imunologia , Leishmania major/imunologia , Infecções por Protozoários/imunologia , Trypanosoma brucei brucei/imunologia , Trypanosoma cruzi/imunologia , Animais , Vesículas Extracelulares/parasitologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Inata/imunologia , Infecções por Protozoários/parasitologia
11.
Nucleic Acids Res ; 49(3): 1436-1454, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33450001

RESUMO

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.


Assuntos
Variação Antigênica , Proteínas de Ligação a DNA/fisiologia , Proteína Homóloga a MRE11/fisiologia , Proteínas de Protozoários/fisiologia , Reparo de DNA por Recombinação , Trypanosoma brucei brucei/genética , Quebras de DNA de Cadeia Dupla , Trypanosoma brucei brucei/imunologia
12.
Sci China Life Sci ; 64(4): 621-632, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33420923

RESUMO

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.


Assuntos
Desoxirribonucleases/imunologia , Armadilhas Extracelulares/imunologia , Evasão da Resposta Imune/imunologia , Proteínas de Protozoários/imunologia , Trypanosoma brucei brucei/imunologia , Trypanosoma/imunologia , Sequência de Aminoácidos , Animais , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Armadilhas Extracelulares/metabolismo , Armadilhas Extracelulares/parasitologia , Feminino , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Varredura , Microscopia Imunoeletrônica , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/parasitologia , Filogenia , Infecções Protozoárias em Animais/imunologia , Infecções Protozoárias em Animais/parasitologia , Proteínas de Protozoários/classificação , Proteínas de Protozoários/metabolismo , Ratos Sprague-Dawley , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Trypanosoma/metabolismo , Trypanosoma/ultraestrutura , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/ultraestrutura
13.
Nat Microbiol ; 6(3): 289-300, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33432154

RESUMO

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.


Assuntos
Splicing de RNA/genética , Transcrição Gênica/genética , Trypanosoma brucei brucei/genética , Glicoproteínas Variantes de Superfície de Trypanosoma/genética , Variação Antigênica/genética , Cromossomos/genética , Cromossomos/metabolismo , Regulação da Expressão Gênica , Genoma de Protozoário/genética , Família Multigênica/genética , RNA Líder para Processamento/genética , Trypanosoma brucei brucei/imunologia
14.
Front Immunol ; 11: 559561, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193328

RESUMO

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.


Assuntos
Doenças dos Bovinos/imunologia , Doenças dos Bovinos/parasitologia , Armadilhas Extracelulares/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/veterinária , Animais , Bovinos , Armadilhas Extracelulares/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Neutrófilos/metabolismo , Consumo de Oxigênio , Espécies Reativas de Oxigênio/metabolismo , Receptores Purinérgicos/metabolismo , Transdução de Sinais
15.
J Biol Chem ; 295(38): 13138-13149, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32727852

RESUMO

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.


Assuntos
Apolipoproteína L1/química , Imunidade Inata , Animais , Apolipoproteína L1/genética , Apolipoproteína L1/imunologia , Sequências Hélice-Alça-Hélice , Humanos , Concentração de Íons de Hidrogênio , Papio hamadryas , Trypanosoma brucei brucei/imunologia
16.
Methods Mol Biol ; 2116: 739-753, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32221952

RESUMO

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.


Assuntos
Linfócitos B/metabolismo , Separação Celular/métodos , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/diagnóstico , Animais , Antígenos CD1d/genética , Antígenos CD1d/metabolismo , Linfócitos B/imunologia , Separação Celular/instrumentação , Modelos Animais de Doenças , Progressão da Doença , Regulação para Baixo/imunologia , Feminino , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Humanos , Separação Imunomagnética/instrumentação , Separação Imunomagnética/métodos , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Camundongos , RNA/isolamento & purificação , RNA-Seq , Receptores de IgE/genética , Receptores de IgE/metabolismo , Baço/citologia , Baço/imunologia , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/parasitologia
17.
Nat Commun ; 11(1): 844, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32051413

RESUMO

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.


Assuntos
Variação Antigênica/genética , Variação Antigênica/imunologia , Recombinação Genética/genética , Trypanosoma vivax/genética , Glicoproteínas Variantes de Superfície de Trypanosoma/genética , Glicoproteínas Variantes de Superfície de Trypanosoma/imunologia , DNA de Protozoário , Evolução Molecular , Genoma de Protozoário , Interações Hospedeiro-Parasita/imunologia , Evasão da Resposta Imune , Filogenia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Homologia de Sequência , Especificidade da Espécie , Transcriptoma , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/parasitologia , Glicoproteínas Variantes de Superfície de Trypanosoma/metabolismo
18.
Proc Natl Acad Sci U S A ; 117(5): 2613-2621, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31964820

RESUMO

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.


Assuntos
Insetos Vetores/parasitologia , Proteínas de Protozoários/genética , Trypanosoma brucei brucei/crescimento & desenvolvimento , Trypanosoma brucei brucei/genética , Moscas Tsé-Tsé/parasitologia , Animais , Feminino , Humanos , Estágios do Ciclo de Vida , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/imunologia , RNA de Protozoário/genética , Glândulas Salivares/parasitologia , Análise de Sequência de RNA , Análise de Célula Única , Transcriptoma , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/parasitologia
19.
Open Biol ; 9(11): 190182, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31718509

RESUMO

African trypanosomes escape the mammalian immune response by antigenic variation-the periodic exchange of one surface coat protein, in Trypanosoma brucei the variant surface glycoprotein (VSG), for an immunologically distinct one. VSG transcription is monoallelic, with only one VSG being expressed at a time from a specialized locus, known as an expression site. VSG switching is a predominantly recombination-driven process that allows VSG sequences to be recombined into the active expression site either replacing the currently active VSG or generating a 'new' VSG by segmental gene conversion. In this review, we describe what is known about the factors that influence this process, focusing specifically on DNA repair and recombination.


Assuntos
Variação Antigênica , Recombinação Genética , Trypanosoma brucei brucei/genética , Tripanossomíase Africana/parasitologia , Glicoproteínas Variantes de Superfície de Trypanosoma/genética , Animais , Reparo do DNA , Conversão Gênica , Humanos , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/veterinária , Glicoproteínas Variantes de Superfície de Trypanosoma/imunologia
20.
Front Immunol ; 10: 2137, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31572363

RESUMO

African trypanosomes, such as Trypanosoma brucei (T. brucei), are protozoan parasites of the mammalian vasculature and central nervous system that are best known for causing fatal human sleeping sickness. As exclusively extracellular parasites, trypanosomes are subject to constant challenge from host immune defenses but they have developed very effective strategies to evade and modulate these responses to maintain an infection while simultaneously prolonging host survival. Here we investigate host parasite interactions, especially within the CNS context, which are not well-understood. We demonstrate that T. brucei strongly upregulates the stress response protein, Heme Oxygenase 1 (HO-1), in primary murine glia and macrophages in vitro. Furthermore, using a novel AHADHinT. brucei cell line, we demonstrate that specific aromatic ketoacids secreted by bloodstream forms of T. brucei are potent drivers of HO-1 expression and are capable of inhibiting pro-IL1ß induction in both glia and macrophages. Additionally, we found that these ketoacids significantly reduced IL-6 and TNFα production by glia, but not macrophages. Finally, we present data to support Nrf2 activation as the mechanism of action by which these ketoacids upregulate HO-1 expression and mediate their anti-inflammatory activity. This study therefore reports a novel immune evasion mechanism, whereby T. brucei secretes amino-acid derived metabolites for the purpose of suppressing both the host CNS and peripheral immune response, potentially via induction of the Nrf2/HO-1 pathway.


Assuntos
Heme Oxigenase-1/imunologia , Macrófagos/imunologia , Proteínas de Membrana/imunologia , Fator 2 Relacionado a NF-E2/imunologia , Neuroglia/imunologia , Piruvatos/imunologia , Trypanosoma brucei brucei/imunologia , Animais , Inflamação/imunologia , Inflamação/patologia , Macrófagos/patologia , Camundongos , Neuroglia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...