Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Cell Mol Life Sci ; 81(1): 134, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38478101

RESUMO

The functions of human Apolipoproteins L (APOLs) are poorly understood, but involve diverse activities like lysis of bloodstream trypanosomes and intracellular bacteria, modulation of viral infection and induction of apoptosis, autophagy, and chronic kidney disease. Based on recent work, I propose that the basic function of APOLs is the control of membrane dynamics, at least in the Golgi and mitochondrion. Together with neuronal calcium sensor-1 (NCS1) and calneuron-1 (CALN1), APOL3 controls the activity of phosphatidylinositol-4-kinase-IIIB (PI4KB), involved in both Golgi and mitochondrion membrane fission. Whereas secreted APOL1 induces African trypanosome lysis through membrane permeabilization of the parasite mitochondrion, intracellular APOL1 conditions non-muscular myosin-2A (NM2A)-mediated transfer of PI4KB and APOL3 from the Golgi to the mitochondrion under conditions interfering with PI4KB-APOL3 interaction, such as APOL1 C-terminal variant expression or virus-induced inflammatory signalling. APOL3 controls mitophagy through complementary interactions with the membrane fission factor PI4KB and the membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). In mice, the basic APOL1 and APOL3 activities could be exerted by mAPOL9 and mAPOL8, respectively. Perspectives regarding the mechanism and treatment of APOL1-related kidney disease are discussed, as well as speculations on additional APOLs functions, such as APOL6 involvement in adipocyte membrane dynamics through interaction with myosin-10 (MYH10).


Assuntos
Apolipoproteína L1 , Insuficiência Renal Crônica , Humanos , Camundongos , Animais , Apolipoproteínas L , Apolipoproteína L1/genética , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Miosinas
2.
Cell Rep ; 42(12): 113528, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38041817

RESUMO

Apolipoproteins L1 and L3 (APOLs) are associated at the Golgi with the membrane fission factors phosphatidylinositol 4-kinase-IIIB (PI4KB) and non-muscular myosin 2A. Either APOL1 C-terminal truncation (APOL1Δ) or APOL3 deletion (APOL3-KO [knockout]) reduces PI4KB activity and triggers actomyosin reorganization. We report that APOL3, but not APOL1, controls PI4KB activity through interaction with PI4KB and neuronal calcium sensor-1 or calneuron-1. Both APOLs are present in Golgi-derived autophagy-related protein 9A vesicles, which are involved in PI4KB trafficking. Like APOL3-KO, APOL1Δ induces PI4KB dissociation from APOL3, linked to reduction of mitophagy flux and production of mitochondrial reactive oxygen species. APOL1 and APOL3, respectively, can interact with the mitophagy receptor prohibitin-2 and the mitophagosome membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). While APOL1 conditions PI4KB and APOL3 involvement in mitochondrion fission and mitophagy, APOL3-VAMP8 interaction promotes fusion between mitophagosomal and endolysosomal membranes. We propose that APOL3 controls mitochondrial membrane dynamics through interactions with the fission factor PI4KB and the fusion factor VAMP8.


Assuntos
Apolipoproteína L1 , Membranas Mitocondriais , Apolipoproteína L1/genética , Membranas Mitocondriais/metabolismo , Complexo de Golgi/metabolismo , Mitocôndrias , 1-Fosfatidilinositol 4-Quinase/metabolismo , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Dinâmica Mitocondrial
3.
Annu Rev Pathol ; 18: 19-45, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36055769

RESUMO

African trypanosomes are bloodstream protozoan parasites that infect mammals including humans, where they cause sleeping sickness. Long-lasting infection is required to favor parasite transmission between hosts. Therefore, trypanosomes have developed strategies to continuously escape innate and adaptive responses of the immune system, while also preventing premature death of the host. The pathology linked to infection mainly results from inflammation and includes anemia and brain dysfunction in addition to loss of specificity and memory of the antibody response. The serum of humans contains an efficient trypanolytic factor, the membrane pore-forming protein apolipoprotein L1 (APOL1). In the two human-infective trypanosomes, specific parasite resistance factors inhibit APOL1 activity. In turn, many African individuals express APOL1 variants that counteract these resistance factors, enabling them to avoid sleeping sickness. However, these variants are associated with chronic kidney disease, particularly in the context of virus-induced inflammation such as coronavirus disease 2019. Vaccination perspectives are discussed.


Assuntos
COVID-19 , Tripanossomíase Africana , Humanos , Apolipoproteína L1/genética , Inflamação , Trypanosoma brucei rhodesiense/fisiologia , Tripanossomíase Africana/genética , Tripanossomíase Africana/parasitologia
4.
Trends Parasitol ; 38(2): 104-108, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34887168

RESUMO

The human serum protein apolipoprotein L1 (APOL1) kills Trypanosoma brucei but not the sleeping sickness agent Trypanosoma rhodesiense. APOL1 C-terminal variants can kill T. rhodesiense but they also induce kidney disease. Given topological and functional differences between intracellular and extracellular APOL1 isoforms, I propose that trypanolysis and kidney disease result from distinct APOL1 activities.


Assuntos
Nefropatias , Podócitos , Trypanosoma , Tripanossomíase Africana , Animais , Apolipoproteína L1/genética , Apolipoproteína L1/metabolismo , Humanos , Nefropatias/metabolismo , Podócitos/metabolismo , Trypanosoma brucei rhodesiense
5.
Curr Opin Immunol ; 72: 13-20, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33721725

RESUMO

Human African trypanosomiasis, or sleeping sickness, results from infection by two subspecies of the protozoan flagellate parasite Trypanosoma brucei, termed Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense, prevalent in western and eastern Africa respectively. These subspecies escape the trypanolytic potential of human serum, which efficiently acts against the prototype species Trypanosoma brucei brucei, responsible for the Nagana disease in cattle. We review the various strategies and components used by trypanosomes to counteract the immune defences of their host, highlighting the adaptive genomic evolution that occurred in both parasite and host to take the lead in this battle. The main parasite surface antigen, named Variant Surface Glycoprotein or VSG, appears to play a key role in different processes involved in the dialogue with the host.


Assuntos
Suscetibilidade a Doenças/imunologia , Predisposição Genética para Doença , Tripanossomíase Africana/etiologia , Imunidade Adaptativa , Apolipoproteína L1/genética , Apolipoproteína L1/metabolismo , Resistência à Doença/genética , Resistência à Doença/imunologia , Regulação da Expressão Gênica , Variação Genética , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Inata , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ligação Proteica , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Trypanosoma brucei gambiense/imunologia , Tripanossomíase Africana/metabolismo
6.
FEBS J ; 288(2): 360-381, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32530132

RESUMO

The discovery that apolipoprotein L1 (APOL1) is the trypanolytic factor of human serum raised interest about the function of APOLs, especially following the unexpected finding that in addition to their protective action against sleeping sickness, APOL1 C-terminal variants also cause kidney disease. Based on the analysis of the structure and trypanolytic activity of APOL1, it was proposed that APOLs could function as ion channels of intracellular membranes and be involved in mechanisms triggering programmed cell death. In this review, the recent finding that APOL1 and APOL3 inversely control the synthesis of phosphatidylinositol-4-phosphate (PI(4)P) by the Golgi PI(4)-kinase IIIB (PI4KB) is commented. APOL3 promotes Ca2+ -dependent activation of PI4KB, but due to their increased interaction with APOL3, APOL1 C-terminal variants can inactivate APOL3, leading to reduction of Golgi PI(4)P synthesis. The impact of APOLs on several pathological processes that depend on Golgi PI(4)P levels is discussed. I propose that through their effect on PI4KB activity, APOLs control not only actomyosin activities related to vesicular trafficking, but also the generation and elongation of autophagosomes induced by inflammation.


Assuntos
Apolipoproteína L1/genética , Apolipoproteínas L/genética , Transtorno Autístico/genética , Neoplasias/genética , Insuficiência Renal/genética , Esquizofrenia/genética , Viroses/genética , Actomiosina/genética , Actomiosina/metabolismo , Animais , Apolipoproteína L1/metabolismo , Apolipoproteínas L/metabolismo , Transtorno Autístico/metabolismo , Transtorno Autístico/patologia , Autofagossomos/metabolismo , Cálcio/metabolismo , Regulação da Expressão Gênica , Complexo de Golgi/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Fosfatos de Fosfatidilinositol/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Esquizofrenia/metabolismo , Esquizofrenia/patologia , Tripanossomíase Africana/tratamento farmacológico , Tripanossomíase Africana/genética , Tripanossomíase Africana/metabolismo , Tripanossomíase Africana/parasitologia , Viroses/metabolismo , Viroses/patologia
8.
iScience ; 23(9): 101476, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32889430

RESUMO

Human innate immunity to Trypanosoma brucei involves the trypanosome C-terminal kinesin TbKIFC1, which transports internalized trypanolytic factor apolipoprotein L1 (APOL1) within the parasite. We show that TbKIFC1 preferentially associates with cholesterol-containing membranes and is indispensable for mammalian infectivity. Knockdown of TbKIFC1 did not affect trypanosome growth in vitro but rendered the parasites unable to infect mice unless antibody synthesis was compromised. Surface clearance of Variant Surface Glycoprotein (VSG)-antibody complexes was far slower in these cells, which were more susceptible to capture by macrophages. This phenotype was not due to defects in VSG expression or trafficking but to decreased VSG mobility in a less fluid, stiffer surface membrane. This change can be attributed to increased cholesterol level in the surface membrane in TbKIFC1 knockdown cells. Clearance of surface-bound antibodies by T. brucei is therefore essential for infectivity and depends on high membrane fluidity maintained by the cholesterol-trafficking activity of TbKIFC1.

9.
ACS Omega ; 5(33): 20953-20959, 2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32875230

RESUMO

A unique feature of the African trypanosome Trypanosoma brucei is the presence of an outer layer made of densely packed variable surface glycoproteins (VSGs), which enables the cells to survive in the bloodstream. Although the VSG coat is critical to pathogenesis, how exactly the glycoproteins are organized at the nanoscale is poorly understood. Here, we show that multiparametric atomic force microscopy is a powerful nanoimaging tool for the structural and mechanical characterization of trypanosomes, in a label-free manner and in buffer solution. Directly correlated images of the structure and elasticity of trypanosomes enable us to identify multiple nanoscale mechanical heterogeneities on the cell surface. On a ∼250 nm scale, regions of softer (Young's modulus ∼50 kPa) and stiffer (∼100 kPa) elasticity alternate, revealing variations of the VSG coat and underlying structures. Our nanoimaging experiments show that the T. brucei cell surface is more heterogeneous than previously anticipated and offer promising prospects for the design of trypanocidal drugs targeting cell surface components.

11.
Cell Rep ; 30(11): 3821-3836.e13, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32187552

RESUMO

The C-terminal variants G1 and G2 of apolipoprotein L1 (APOL1) confer human resistance to the sleeping sickness parasite Trypanosoma rhodesiense, but they also increase the risk of kidney disease. APOL1 and APOL3 are death-promoting proteins that are partially associated with the endoplasmic reticulum and Golgi membranes. We report that in podocytes, either APOL1 C-terminal helix truncation (APOL1Δ) or APOL3 deletion (APOL3KO) induces similar actomyosin reorganization linked to the inhibition of phosphatidylinositol-4-phosphate [PI(4)P] synthesis by the Golgi PI(4)-kinase IIIB (PI4KB). Both APOL1 and APOL3 can form K+ channels, but only APOL3 exhibits Ca2+-dependent binding of high affinity to neuronal calcium sensor-1 (NCS-1), promoting NCS-1-PI4KB interaction and stimulating PI4KB activity. Alteration of the APOL1 C-terminal helix triggers APOL1 unfolding and increased binding to APOL3, affecting APOL3-NCS-1 interaction. Since the podocytes of G1 and G2 patients exhibit an APOL1Δ or APOL3KO-like phenotype, APOL1 C-terminal variants may induce kidney disease by preventing APOL3 from activating PI4KB, with consecutive actomyosin reorganization of podocytes.


Assuntos
Actomiosina/metabolismo , Apolipoproteína L1/química , Apolipoproteína L1/genética , Apolipoproteínas L/metabolismo , Nefropatias/metabolismo , Mutação/genética , Sequência de Aminoácidos , Apolipoproteína L1/urina , Cálcio/metabolismo , Linhagem Celular , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Humanos , Nefropatias/urina , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Antígenos de Histocompatibilidade Menor/metabolismo , Proteínas Sensoras de Cálcio Neuronal/metabolismo , Neuropeptídeos/metabolismo , Fenótipo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Podócitos/ultraestrutura , Poli I-C/farmacologia , Canais de Potássio/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína
12.
Microbiol Res ; 205: 73-79, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28942847

RESUMO

Trypanosoma cruzi, the etiological agent of the Chagas' disease in Latin America undergoes a complex life cycle involving two hosts, a mammalian host and a reduviid insect vector (triatomine). In the insect midgut the parasite multiplies as epimastigote forms, which rely on endocytosis for their energy requirement. We recently showed that posttranslational modification of endocytic N-glycoproteins by tomato lectin (TL) binding-N-glycans is crucial for receptor-mediated endocytosis (RME) in epimastigote forms. In an attempt to characterize the endocytic proteome we used a TL affinity chromatography, which significantly enriched glycoproteins of the trypanosomal endocytic pathway. In addition to various lysosomal hydrolases, we found an endosomal C-type lectin-like protein, which displays some structural and topological characteristics of the mammalian lectin receptor superfamily. This lectin encoding a large transmembrane protein of around 375kDa contained three putative extracellular N-terminal C-type lectin domains (CTLD) and located inside the flagellar pocket (FP)/cytostome and endosomal compartments of the insect stage of the parasite and on the surface of the plasma membrane of intracellular amastigote parasites. Noteworthy, this endogenous lectin displayed similar sugar-binding specificity to that of TL and therefore could be important in either the N-glycan mediated endocytosis or parasite adhesion to host cells. We postulated that during the evolution of trypanosomatids, genes encoding lectin harboring 3 CTDLs represent an old acquisition present in free-living, monoxenic and heteroxenic trypanosomatids, which would have been secondarily lost in extracellular parasites from the T. brucei clade.


Assuntos
Lectinas Tipo C/genética , Lectinas Tipo C/isolamento & purificação , Lectinas de Plantas , Proteoma/metabolismo , Trypanosoma cruzi/imunologia , Trypanosoma cruzi/metabolismo , Animais , Endocitose/fisiologia , Humanos , Lectinas Tipo C/classificação , Filogenia , Domínios e Motivos de Interação entre Proteínas , Proteínas de Protozoários/genética , Proteínas Recombinantes , Alinhamento de Sequência , Trypanosoma cruzi/genética
13.
Nat Microbiol ; 2(11): 1500-1506, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28924146

RESUMO

The primate-specific serum protein apolipoprotein L1 (APOL1) is the only secreted member of a family of cell death promoting proteins 1-4 . APOL1 kills the bloodstream parasite Trypanosoma brucei brucei, but not the human sleeping sickness agents T.b. rhodesiense and T.b. gambiense 3 . We considered the possibility that intracellular members of the APOL1 family, against which extracellular trypanosomes could not have evolved resistance, could kill pathogenic T. brucei subspecies. Here we show that recombinant APOL3 (rAPOL3) kills all African trypanosomes, including T.b. rhodesiense, T.b. gambiense and the animal pathogens Trypanosoma evansi, Trypanosoma congolense and Trypanosoma vivax. However, rAPOL3 did not kill more distant trypanosomes such as Trypanosoma theileri or Trypanosoma cruzi. This trypanolytic potential was partially shared by rAPOL1 from Papio papio (rPpAPOL1). The differential killing ability of rAPOL3 and rAPOL1 was associated with a distinct dependence on acidic pH for activity. Due both to its instability and toxicity when injected into mice, rAPOL3 cannot be used for the treatment of infection, but an experimental rPpAPOL1 mutant inspired by APOL3 exhibited enhanced trypanolytic activity in vitro and the ability to completely inhibit T.b. gambiense infection in mice. We conclude that pH dependence influences the trypanolytic potential of rAPOLs.


Assuntos
Apolipoproteína L1/farmacologia , Apolipoproteínas L/farmacologia , Trypanosoma/efeitos dos fármacos , Animais , Apolipoproteína L1/genética , Apolipoproteínas L/genética , Concentração de Íons de Hidrogênio , Camundongos , Papio papio , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/farmacologia , Trypanosoma/fisiologia , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma brucei gambiense/efeitos dos fármacos , Trypanosoma brucei rhodesiense/efeitos dos fármacos , Trypanosoma congolense/efeitos dos fármacos , Trypanosoma vivax/efeitos dos fármacos , Tripanossomíase Africana/parasitologia
14.
Sci Rep ; 7(1): 6165, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28733685

RESUMO

Infection of C57Bl/6 mice by pleomorphic African trypanosomes Trypanosoma brucei and T. congolense is characterized by parasitemia waves coupled with the production of systemic levels of TNF. This cytokine is known to control T. brucei growth, but also to contribute to tissue damage, shortening the survival time of infected mice. Using a dominant-negative version of TNF to discriminate between the effects of the membrane-form versus the soluble form of TNF, we show that the second form is involved in neither parasite control nor induction of liver injury. Therefore, soluble TNF is likely not a major contributor to disease outcome. We propose that membrane-bound TNF is responsible for both T. brucei control and host pathology.


Assuntos
Parasitemia/veterinária , Trypanosoma brucei brucei/crescimento & desenvolvimento , Tripanossomíase Africana/parasitologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Membrana Celular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/veterinária , Fator de Necrose Tumoral alfa/farmacologia
15.
J Immunol ; 199(5): 1762-1771, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28739879

RESUMO

Arginase activity induction in macrophages is an escape mechanism developed by parasites to cope with the host's immune defense and benefit from increased host-derived growth factor production. We report that arginase expression and activity were induced in macrophages during mouse infection by Trypanosoma musculi, a natural parasite of this host. This induction was reproduced in vitro by excreted/secreted factors of the parasite. A mAb directed to TbKHC1, an orphan kinesin H chain from Trypanosoma brucei, inhibited T. musculi excreted/secreted factor-mediated arginase induction. Anti-TbKHC1 Ab also inhibited T. musculi growth, both in vitro and in vivo. Induction of arginase activity and parasite growth involved C-type lectin receptors, because mannose injection decreased arginase activity induction and parasite load in vitro and in vivo. Accordingly, the parasite load was reduced in mice lacking mannose receptor C-type 1. The T. musculi KHC1 homolog showed high similarity with TbKHC1. Bioinformatics analysis revealed the presence of homologs of this gene in other trypanosomes, including pathogens for humans and animals. Host metabolism dysregulation represents an effective parasite mechanism to hamper the host immune response and modify host molecule production to favor parasite invasion and growth. Thus, this orphan kinesin plays an important role in promoting trypanosome infection, and its neutralization or the lock of its partner host molecules offers promising approaches to increasing resistance to infection and new developments in vaccination against trypanosomiasis.


Assuntos
Antígenos de Protozoários/metabolismo , Arginase/metabolismo , Moléculas de Adesão Celular/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/imunologia , Receptores de Superfície Celular/metabolismo , Trypanosoma/fisiologia , Tripanossomíase/imunologia , Animais , Anticorpos/metabolismo , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Moléculas de Adesão Celular/genética , Células Cultivadas , Feminino , Cinesinas/genética , Lectinas Tipo C/genética , Macrófagos/parasitologia , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Carga Parasitária , Filogenia , Receptores de Superfície Celular/genética , Vacinação
16.
PLoS One ; 11(9): e0163302, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27685262

RESUMO

Trypanosoma cruzi is a protozoan parasite transmitted by a triatomine insect, and causing human Chagas disease in South America. This parasite undergoes a complex life cycle alternating between non-proliferative and dividing forms. Owing to their high energy requirement, replicative epimastigotes of the insect midgut display high endocytic activity. This activity is mainly restricted to the cytostome, by which the cargo is taken up and sorted through the endosomal vesicular network to be delivered to reservosomes, the final lysosomal-like compartments. In African trypanosomes tomato lectin (TL) and ricin, respectively specific to poly-N-acetyllactosamine (poly-LacNAc) and ß-D-galactose, allowed the identification of giant chains of poly-LacNAc in N-glycoproteins of the endocytic pathway. We show that in T. cruzi epimastigote forms also, glycoproteins of the endocytic pathway are characterized by the presence of N-linked glycans binding to both ricin and TL. Affinity chromatography using both TL and Griffonia simplicifolia lectin II (GSLII), specific to non-reducing terminal residue of N-acetylglucosamine (GlcNAc), led to an enrichment of glycoproteins of the trypanosomal endocytic pathway. Incubation of live parasites with TL, which selectively bound to the cytostome/cytopharynx, specifically inhibited endocytosis of transferrin (Tf) but not dextran, a marker of fluid endocytosis. Taken together, our data suggest that N-glycan modification of endocytic components plays a crucial role in receptor-mediated endocytosis of T. cruzi.

17.
Eur J Immunol ; 46(8): 1854-66, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27198486

RESUMO

Apolipoproteins L (ApoLs) are Bcl-2-like proteins expressed under inflammatory conditions in myeloid and endothelial cells. We found that Toll-like receptor (TLR) stimuli, particularly the viral mimetic polyinosinic:polycytidylic acid (poly(I:C)), specifically induce ApoLs7/11 subfamilies in murine CD8α(+)  dendritic cells (DCs). This induction requires the TLR3/TRIF (where TRIF is TIR domain containing adapter-inducing interferon ß) signaling pathway and is dependent on IFN-ß in all ApoLs subfamilies except for ApoL7c. Poly(I:C) treatment of DCs is also associated with induction of both cell death and autophagy. ApoLs expression is related to promotion of DC death by poly(I:C), as ApoLs7/11 knockdown increases DC survival and ApoLs7 are associated with the anti-apoptotic protein Bcl-xL (where Bcl-xL is B-cell lymphoma extra large). Similarly, in human monocyte-derived DCs poly(I:C) induces both cell death and the expression of ApoLs, principally ApoL3. Finally, the BH3-like peptide of ApoLs appears to be involved in the DC death-promoting activity. We would like to propose that ApoLs are involved in cell death linked to activation of DCs by viral stimuli.


Assuntos
Apolipoproteínas/imunologia , Apoptose , Células Dendríticas/citologia , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antígenos CD8/metabolismo , Linhagem Celular , Células Cultivadas , Células Dendríticas/metabolismo , Humanos , Interferon beta/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli I-C/farmacologia , Isoformas de Proteínas/imunologia , Proteína bcl-X/metabolismo
18.
mBio ; 7(2): e02198-15, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27073096

RESUMO

UNLABELLED: African trypanosomes, except Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense, which cause human African trypanosomiasis, are lysed by the human serum protein apolipoprotein L1 (ApoL1). These two subspecies can resist human ApoL1 because they express the serum resistance proteins T. b. gambiense glycoprotein (TgsGP) and serum resistance-associated protein (SRA), respectively. Whereas in T. b. rhodesiense, SRA is necessary and sufficient to inhibit ApoL1, in T. b. gambiense, TgsGP cannot protect against high ApoL1 uptake, so different additional mechanisms contribute to limit this uptake. Here we report a complex interplay between trypanosomes and an ApoL1 variant, revealing important insights into innate human immunity against these parasites. Using whole-genome sequencing, we characterized an atypical T. b. gambiense infection in a patient in Ghana. We show that the infecting trypanosome has diverged from the classical T. b. gambiense strains and lacks the TgsGP defense mechanism against human serum. By sequencing the ApoL1 gene of the patient and subsequent in vitro mutagenesis experiments, we demonstrate that a homozygous missense substitution (N264K) in the membrane-addressing domain of this ApoL1 variant knocks down the trypanolytic activity, allowing the trypanosome to avoid ApoL1-mediated immunity. IMPORTANCE: Most African trypanosomes are lysed by the ApoL1 protein in human serum. Only the subspecies Trypanosoma b. gambiense and T. b. rhodesiense can resist lysis by ApoL1 because they express specific serum resistance proteins. We here report a complex interplay between trypanosomes and an ApoL1 variant characterized by a homozygous missense substitution (N264K) in the domain that we hypothesize interacts with the endolysosomal membranes of trypanosomes. The N264K substitution knocks down the lytic activity of ApoL1 against T. b. gambiense strains lacking the TgsGP defense mechanism and against T. b. rhodesiense if N264K is accompanied by additional substitutions in the SRA-interacting domain. Our data suggest that populations with high frequencies of the homozygous N264K ApoL1 variant may be at increased risk of contracting human African trypanosomiasis.


Assuntos
Apolipoproteínas/genética , Suscetibilidade a Doenças , Variação Genética , Lipoproteínas HDL/genética , Trypanosoma brucei gambiense/fisiologia , Trypanosoma brucei rhodesiense/fisiologia , Tripanossomíase Africana/genética , Apolipoproteína L1 , Apolipoproteínas/imunologia , Humanos , Imunidade Inata , Lipoproteínas HDL/imunologia , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/parasitologia
19.
Nat Commun ; 6: 8078, 2015 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-26307671

RESUMO

Humans resist infection by the African parasite Trypanosoma brucei owing to the trypanolytic activity of the serum apolipoprotein L1 (APOL1). Following uptake by endocytosis in the parasite, APOL1 forms pores in endolysosomal membranes and triggers lysosome swelling. Here we show that APOL1 induces both lysosomal and mitochondrial membrane permeabilization (LMP and MMP). Trypanolysis coincides with MMP and consecutive release of the mitochondrial TbEndoG endonuclease to the nucleus. APOL1 is associated with the kinesin TbKIFC1, of which both the motor and vesicular trafficking VHS domains are required for MMP, but not for LMP. The presence of APOL1 in the mitochondrion is accompanied by mitochondrial membrane fenestration, which can be mimicked by knockdown of a mitochondrial mitofusin-like protein (TbMFNL). The BH3-like peptide of APOL1 is required for LMP, MMP and trypanolysis. Thus, trypanolysis by APOL1 is linked to apoptosis-like MMP occurring together with TbKIFC1-mediated transport of APOL1 from endolysosomal membranes to the mitochondrion.


Assuntos
Apolipoproteínas/metabolismo , Cinesinas/metabolismo , Lipoproteínas HDL/metabolismo , Lisossomos/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas de Protozoários/metabolismo , Apolipoproteína L1 , Apoptose , Transporte Biológico , Endocitose , Humanos , Membranas Intracelulares/metabolismo , Permeabilidade , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/patogenicidade , Trypanosoma brucei gambiense/metabolismo , Trypanosoma brucei gambiense/patogenicidade , Trypanosoma brucei rhodesiense/metabolismo , Trypanosoma brucei rhodesiense/patogenicidade
20.
Kidney Int ; 88(4): 754-63, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25993319

RESUMO

A third of African Americans with sporadic focal segmental glomerulosclerosis (FSGS) or HIV-associated nephropathy (HIVAN) do not carry APOL1 renal risk genotypes. This raises the possibility that other APOL1 variants may contribute to kidney disease. To address this question, we sequenced all APOL1 exons in 1437 Americans of African and European descent, including 464 patients with biopsy-proven FSGS/HIVAN. Testing for association with 33 common and rare variants with FSGS/HIVAN revealed no association independent of strong recessive G1 and G2 effects. Seeking additional variants that might have been under selection by pathogens and could represent candidates for kidney disease risk, we also sequenced an additional 1112 individuals representing 53 global populations. Except for G1 and G2, none of the 7 common codon-altering variants showed evidence of selection or could restore lysis against trypanosomes causing human African trypanosomiasis. Thus, only APOL1 G1 and G2 confer renal risk, and other common and rare APOL1 missense variants, including the archaic G3 haplotype, do not contribute to sporadic FSGS and HIVAN in the US population. Hence, in most potential clinical or screening applications, our study suggests that sequencing APOL1 exons is unlikely to bring additional information compared to genotyping only APOL1 G1 and G2 risk alleles.


Assuntos
Nefropatia Associada a AIDS/genética , Apolipoproteínas/genética , Glomerulosclerose Segmentar e Focal/genética , Lipoproteínas HDL/genética , Polimorfismo de Nucleotídeo Único , Nefropatia Associada a AIDS/diagnóstico , Nefropatia Associada a AIDS/etnologia , Negro ou Afro-Americano/genética , Apolipoproteína L1 , Apolipoproteínas/sangue , Biópsia , Estudos de Casos e Controles , Éxons , Feminino , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Glomerulosclerose Segmentar e Focal/diagnóstico , Glomerulosclerose Segmentar e Focal/etnologia , Haplótipos , Interações Hospedeiro-Parasita , Humanos , Lipoproteínas HDL/sangue , Masculino , Fenótipo , Medição de Risco , Fatores de Risco , Análise de Sequência de DNA , Trypanosoma brucei gambiense/metabolismo , Trypanosoma brucei gambiense/patogenicidade , Trypanosoma brucei rhodesiense/metabolismo , Trypanosoma brucei rhodesiense/patogenicidade , Estados Unidos/epidemiologia , População Branca/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...