Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 134, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38478101

RESUMEN

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).


Asunto(s)
Apolipoproteína L1 , Insuficiencia Renal Crónica , Humanos , Ratones , Animales , Apolipoproteínas L , Apolipoproteína L1/genética , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Miosinas
2.
EMBO J ; 34(10): 1336-48, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25787857

RESUMEN

The severity and intensity of autoimmune disease in immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) patients and in scurfy mice emphasize the critical role played by thymus-derived regulatory T cells (tTregs) in maintaining peripheral immune tolerance. However, although tTregs are critical to prevent lethal autoimmunity and excessive inflammatory responses, their suppressive mechanism remains elusive. Here, we demonstrate that tTregs selectively inhibit CD27/CD70-dependent Th1 priming, while leaving the IL-12-dependent pathway unaffected. Immunized mice depleted of tTregs showed an increased response of IFN-γ-secreting CD4(+) T cells that was strictly reliant on a functional CD27/CD70 pathway. In vitro studies revealed that tTregs downregulate CD70 from the plasma membrane of dendritic cells (DCs) in a CD27-dependent manner. CD70 downregulation required contact between Tregs and DCs and resulted in endocytosis of CD27 and CD70 into the DC. These findings reveal a novel mechanism by which tTregs can maintain tolerance or prevent excessive, proinflammatory Th1 responses.


Asunto(s)
Ligando CD27/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Timo/inmunología , Timo/metabolismo , Animales , Ligando CD27/genética , Células Dendríticas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
3.
J Immunol ; 199(5): 1762-1771, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28739879

RESUMEN

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.


Asunto(s)
Antígenos de Protozoos/metabolismo , Arginasa/metabolismo , Moléculas de Adhesión Celular/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/inmunología , Receptores de Superficie Celular/metabolismo , Trypanosoma/fisiología , Tripanosomiasis/inmunología , Animales , Anticuerpos/metabolismo , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Moléculas de Adhesión Celular/genética , Células Cultivadas , Femenino , Cinesinas/genética , Lectinas Tipo C/genética , Macrófagos/parasitología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Carga de Parásitos , Filogenia , Receptores de Superficie Celular/genética , Vacunación
4.
Nature ; 501(7467): 430-4, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23965626

RESUMEN

The African parasite Trypanosoma brucei gambiense accounts for 97% of human sleeping sickness cases. T. b. gambiense resists the specific human innate immunity acting against several other tsetse-fly-transmitted trypanosome species such as T. b. brucei, the causative agent of nagana disease in cattle. Human immunity to some African trypanosomes is due to two serum complexes designated trypanolytic factors (TLF-1 and -2), which both contain haptoglobin-related protein (HPR) and apolipoprotein LI (APOL1). Whereas HPR association with haemoglobin (Hb) allows TLF-1 binding and uptake via the trypanosome receptor TbHpHbR (ref. 5), TLF-2 enters trypanosomes independently of TbHpHbR (refs 4, 5). APOL1 kills trypanosomes after insertion into endosomal/lysosomal membranes. Here we report that T. b. gambiense resists TLFs via a hydrophobic ß-sheet of the T. b. gambiense-specific glycoprotein (TgsGP), which prevents APOL1 toxicity and induces stiffening of membranes upon interaction with lipids. Two additional features contribute to resistance to TLFs: reduction of sensitivity to APOL1 requiring cysteine protease activity, and TbHpHbR inactivation due to a L210S substitution. According to such a multifactorial defence mechanism, transgenic expression of T. b. brucei TbHpHbR in T. b. gambiense did not cause parasite lysis in normal human serum. However, these transgenic parasites were killed in hypohaptoglobinaemic serum, after high TLF-1 uptake in the absence of haptoglobin (Hp) that competes for Hb and receptor binding. TbHpHbR inactivation preventing high APOL1 loading in hypohaptoglobinaemic serum may have evolved because of the overlapping endemic area of T. b. gambiense infection and malaria, the main cause of haemolysis-induced hypohaptoglobinaemia in western and central Africa.


Asunto(s)
Apolipoproteínas/sangre , Apolipoproteínas/metabolismo , Lipoproteínas HDL/sangre , Lipoproteínas HDL/metabolismo , Trypanosoma brucei gambiense/fisiología , África , Animales , Animales Modificados Genéticamente , Apolipoproteína L1 , Apolipoproteínas/antagonistas & inhibidores , Apolipoproteínas/toxicidad , Membrana Celular/química , Membrana Celular/metabolismo , Proteasas de Cisteína/metabolismo , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Hemólisis , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Metabolismo de los Lípidos , Lipoproteínas HDL/antagonistas & inhibidores , Lipoproteínas HDL/química , Lipoproteínas HDL/toxicidad , Parásitos/patogenicidad , Parásitos/fisiología , Estructura Secundaria de Proteína , Suero/química , Suero/parasitología , Trypanosoma brucei gambiense/efectos de los fármacos , Trypanosoma brucei gambiense/patogenicidad , Tripanosomiasis Africana/parasitología , Glicoproteínas Variantes de Superficie de Trypanosoma/química , Glicoproteínas Variantes de Superficie de Trypanosoma/metabolismo
5.
Eur J Immunol ; 46(8): 1854-66, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27198486

RESUMEN

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.


Asunto(s)
Apolipoproteínas/inmunología , Apoptosis , Células Dendríticas/citología , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígenos CD8/metabolismo , Línea Celular , Células Cultivadas , Células Dendríticas/metabolismo , Humanos , Interferón beta/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/farmacología , Isoformas de Proteínas/inmunología , Proteína bcl-X/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(24): 8943-8, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24889641

RESUMEN

African trypanosomes survive the immune defense of their hosts by regularly changing their antigenic coat made of variant surface glycoprotein (VSG). The Trypanosoma brucei genome contains more than 1,000 VSG genes. To be expressed, a given VSG gene must be located in one of 15 telomeric regions termed "VSG expression sites" (ESs), each of which contains a polycistronic transcription unit that includes ES-associated genes. Only one ES is fully active at a time, so only one VSG gene is transcribed per cell. Although this monoallelic expression is controlled at the transcriptional level, the precise molecular mechanism for this control is not understood. Here we report that in single cells transcription is initiated on several ESs simultaneously, indicating that the monoallelic control is not determined only at transcription initiation, but also at further control steps such as transcription elongation or RNA processing.


Asunto(s)
Transcripción Genética , Trypanosoma brucei brucei/metabolismo , Glicoproteínas Variantes de Superficie de Trypanosoma/genética , Alelos , Variación Antigénica , Línea Celular , Cartilla de ADN , Genes Protozoarios , Variación Genética , Humanos , Reacción en Cadena de la Polimerasa , Tripanosomiasis Africana/sangre , Tripanosomiasis Africana/parasitología , Glicoproteínas Variantes de Superficie de Trypanosoma/metabolismo
7.
Mol Microbiol ; 97(3): 397-407, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25899052

RESUMEN

Human apolipoprotein L1 (APOL1) kills African trypanosomes except Trypanosoma rhodesiense and Trypanosoma gambiense, the parasites causing sleeping sickness. APOL1 uptake into trypanosomes is favoured by its association with the haptoglobin-related protein-haemoglobin complex, which binds to the parasite surface receptor for haptoglobin-haemoglobin. As haptoglobin-haemoglobin can saturate the receptor, APOL1 uptake is increased in haptoglobin-poor (hypohaptoglobinaemic) serum (HyHS). While T. rhodesiense resists APOL1 by RNA polymerase I (pol-I)-mediated expression of the serum resistance-associated (SRA) protein, T. gambiense resists by pol-II-mediated expression of the T. gambiense-specific glycoprotein (TgsGP). Moreover, in T. gambiense resistance to HyHS is linked to haptoglobin-haemoglobin receptor inactivation by mutation. We report that unlike T. gambiense, T. rhodesiense possesses a functional haptoglobin-haemoglobin receptor, and that like T. gambiense experimentally provided with active receptor, this parasite is killed in HyHS because of receptor-mediated APOL1 uptake. However, T. rhodesiense could adapt to low haptoglobin by increasing transcription of SRA. When assayed in Trypanosoma brucei, resistance to HyHS occurred with pol-I-, but not with pol-II-mediated SRA expression. Similarly, T. gambiense provided with active receptor acquired resistance to HyHS only when TgsGP was moved to a pol-I locus. Thus, transcription by pol-I favours adaptive gene regulation, explaining the presence of SRA in a pol-I locus.


Asunto(s)
Apolipoproteínas/toxicidad , Regulación de la Expresión Génica , Lipoproteínas HDL/toxicidad , ARN Polimerasa I/metabolismo , Transcripción Genética , Trypanosoma brucei rhodesiense/efectos de los fármacos , Trypanosoma brucei rhodesiense/fisiología , Adaptación Fisiológica , Apolipoproteína L1 , Haptoglobinas/análisis , Humanos , Glicoproteínas de Membrana/biosíntesis , Receptores de Superficie Celular/metabolismo , Suero/química , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/crecimiento & desarrollo , Trypanosoma brucei gambiense/efectos de los fármacos , Trypanosoma brucei gambiense/genética , Trypanosoma brucei gambiense/crecimiento & desarrollo , Trypanosoma brucei rhodesiense/genética , Trypanosoma brucei rhodesiense/crecimiento & desarrollo
10.
Kidney Int ; 88(4): 754-63, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25993319

RESUMEN

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.


Asunto(s)
Nefropatía Asociada a SIDA/genética , Apolipoproteínas/genética , Glomeruloesclerosis Focal y Segmentaria/genética , Lipoproteínas HDL/genética , Polimorfismo de Nucleótido Simple , Nefropatía Asociada a SIDA/diagnóstico , Nefropatía Asociada a SIDA/etnología , Negro o Afroamericano/genética , Apolipoproteína L1 , Apolipoproteínas/sangre , Biopsia , Estudios de Casos y Controles , Exones , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Glomeruloesclerosis Focal y Segmentaria/etnología , Haplotipos , Interacciones Huésped-Parásitos , Humanos , Lipoproteínas HDL/sangre , Masculino , Fenotipo , Medición de Riesgo , Factores de Riesgo , Análisis de Secuencia de ADN , Trypanosoma brucei gambiense/metabolismo , Trypanosoma brucei gambiense/patogenicidad , Trypanosoma brucei rhodesiense/metabolismo , Trypanosoma brucei rhodesiense/patogenicidad , Estados Unidos/epidemiología , Población Blanca/genética
11.
Mol Microbiol ; 94(3): 625-36, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25256834

RESUMEN

Normal human serum (NHS) confers human resistance to infection by the parasite Trypanosoma brucei owing to the trypanolytic activity of apolipoprotein L1 (APOL1), present in two serum complexes termed Trypanolytic Factors (TLF-1 and -2). In order to identify parasite components involved in the intracellular trafficking and activity of TLFs, an inducible RNA interference (RNAi) genomic DNA library constructed in bloodstream form T. brucei was subjected to RNAi induction and selection for resistant parasites under NHS conditions favouring either TLF-1 or TLF-2 uptake. While TLF-1 conditions readily selected the haptoglobin-haemoglobin (HP-HB) surface receptor TbHpHbR as expected, given its known ability to bind TLF-1, under TLF-2 conditions no specific receptor for TLF-2 was identified. Instead, the screen allowed the identification of five distinct factors expected to be involved in the assembly of the vacuolar proton pump V-ATPase and consecutive endosomal acidification. These data confirm that lowering the pH during endocytosis is required for APOL1 toxic activity.


Asunto(s)
Apolipoproteínas/metabolismo , Citotoxinas/metabolismo , Lipoproteínas HDL/metabolismo , Suero/metabolismo , Trypanosoma brucei brucei/efectos de los fármacos , Apolipoproteína L1 , Endocitosis , Concentración de Iones de Hidrógeno , Proteínas Protozoarias/análisis , Proteínas Protozoarias/genética , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/genética , Trypanosoma brucei brucei/genética
12.
PLoS Pathog ; 9(10): e1003731, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204274

RESUMEN

BACKGROUND: In order to promote infection, the blood-borne parasite Trypanosoma brucei releases factors that upregulate arginase expression and activity in myeloid cells. METHODOLOGY/PRINCIPAL FINDINGS: By screening a cDNA library of T. brucei with an antibody neutralizing the arginase-inducing activity of parasite released factors, we identified a Kinesin Heavy Chain isoform, termed TbKHC1, as responsible for this effect. Following interaction with mouse myeloid cells, natural or recombinant TbKHC1 triggered SIGN-R1 receptor-dependent induction of IL-10 production, resulting in arginase-1 activation concomitant with reduction of nitric oxide (NO) synthase activity. This TbKHC1 activity was IL-4Rα-independent and did not mirror M2 activation of myeloid cells. As compared to wild-type T. brucei, infection by TbKHC1 KO parasites was characterized by strongly reduced parasitaemia and prolonged host survival time. By treating infected mice with ornithine or with NO synthase inhibitor, we observed that during the first wave of parasitaemia the parasite growth-promoting effect of TbKHC1-mediated arginase activation resulted more from increased polyamine production than from reduction of NO synthesis. In late stage infection, TbKHC1-mediated reduction of NO synthesis appeared to contribute to liver damage linked to shortening of host survival time. CONCLUSION: A kinesin heavy chain released by T. brucei induces IL-10 and arginase-1 through SIGN-R1 signaling in myeloid cells, which promotes early trypanosome growth and favors parasite settlement in the host. Moreover, in the late stage of infection, the inhibition of NO synthesis by TbKHC1 contributes to liver pathogenicity.


Asunto(s)
Arginasa/inmunología , Cinesinas/inmunología , Proteínas Protozoarias/inmunología , Trypanosoma brucei brucei/inmunología , Tripanosomiasis Africana/inmunología , Animales , Arginasa/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Activación Enzimática/genética , Activación Enzimática/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Cinesinas/genética , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ratones , Ratones Noqueados , Óxido Nítrico/genética , Óxido Nítrico/inmunología , Proteínas Protozoarias/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Trypanosoma brucei brucei/genética , Tripanosomiasis Africana/genética , Tripanosomiasis Africana/patología
13.
Eukaryot Cell ; 12(2): 168-81, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23104568

RESUMEN

FKBP12 proteins are able to inhibit TOR kinases or calcineurin phosphatases upon binding of rapamycin or FK506 drugs, respectively. The Trypanosoma brucei FKBP12 homologue (TbFKBP12) was found to be a cytoskeleton-associated protein with specific localization in the flagellar pocket area of the bloodstream form. In the insect procyclic form, RNA interference-mediated knockdown of TbFKBP12 affected motility. In bloodstream cells, depletion of TbFKBP12 affected cytokinesis and cytoskeleton architecture. These last effects were associated with the presence of internal translucent cavities limited by an inside-out configuration of the normal cell surface, with a luminal variant surface glycoprotein coat lined up by microtubules. These cavities, which recreated the streamlined shape of the normal trypanosome cytoskeleton, might represent unsuccessful attempts for cell abscission. We propose that TbFKBP12 differentially affects stage-specific processes through association with the cytoskeleton.


Asunto(s)
Citocinesis , Proteínas Protozoarias/fisiología , Proteína 1A de Unión a Tacrolimus/metabolismo , Trypanosoma brucei brucei/enzimología , Movimiento Celular , ADN de Cinetoplasto/metabolismo , ADN Protozoario/metabolismo , Flagelos/metabolismo , Flagelos/ultraestructura , Técnicas de Silenciamiento del Gen , Microtúbulos/metabolismo , Transporte de Proteínas , Proteínas Protozoarias/genética , ARN Interferente Pequeño/genética , Proteína 1A de Unión a Tacrolimus/genética , Trypanosoma brucei brucei/crecimiento & desarrollo , Trypanosoma brucei brucei/ultraestructura
14.
Mol Microbiol ; 84(2): 225-42, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22340731

RESUMEN

Antigenic variation of the parasite Trypanosoma brucei operates by monoallelic expression of a variant surface glycoprotein (VSG) from a collection of multiple telomeric expression sites (ESs). Each of these ESs harbours a long polycistronic transcription unit containing several expression site-associated genes (ESAGs). ESAG4 copies encode bloodstream stage-specific adenylyl cyclases (AC) and belong to a larger gene family of around 80 members, the majority of which, termed genes related to ESAG4 (GRESAG4s), are not encoded in ESs and are expressed constitutively in the life cycle. Here we report that ablation of ESAG4 from the active ES did not affect parasite growth, neither in culture nor upon rodent infection, and did not significantly change total AC activity. In contrast, inducible RNAi-mediated knock-down of an AC subfamily that includes ESAG4 and two ESAG4-like GRESAG4 (ESAG4L) genes, decreased total AC activity and induced a lethal phenotype linked to impaired cytokinesis. In the Δesag4 line compensatory upregulation of apparently functionally redundant ESAG4L genes was observed, suggesting that the ESAG4/ESAG4L-subfamily ACs are involved in the control of cell division. How deregulated adenylyl cyclases or cAMP might impair cytokinesis is discussed.


Asunto(s)
Adenilil Ciclasas/metabolismo , Citocinesis , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/fisiología , Adenilil Ciclasas/genética , Análisis por Conglomerados , Eliminación de Gen , Expresión Génica , Prueba de Complementación Genética , Filogenia , Proteínas Protozoarias/genética , Homología de Secuencia de Aminoácido , Trypanosoma brucei brucei/genética
15.
PLoS Pathog ; 7(6): e1002072, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21698216

RESUMEN

The African trypanosome Trypanosoma brucei, which persists within the bloodstream of the mammalian host, has evolved potent mechanisms for immune evasion. Specifically, antigenic variation of the variant-specific surface glycoprotein (VSG) and a highly active endocytosis and recycling of the surface coat efficiently delay killing mediated by anti-VSG antibodies. Consequently, conventional VSG-specific intact immunoglobulins are non-trypanocidal in the absence of complement. In sharp contrast, monovalent antigen-binding fragments, including 15 kDa nanobodies (Nb) derived from camelid heavy-chain antibodies (HCAbs) recognizing variant-specific VSG epitopes, efficiently lyse trypanosomes both in vitro and in vivo. This Nb-mediated lysis is preceded by very rapid immobilisation of the parasites, massive enlargement of the flagellar pocket and major blockade of endocytosis. This is accompanied by severe metabolic perturbations reflected by reduced intracellular ATP-levels and loss of mitochondrial membrane potential, culminating in cell death. Modification of anti-VSG Nbs through site-directed mutagenesis and by reconstitution into HCAbs, combined with unveiling of trypanolytic activity from intact immunoglobulins by papain proteolysis, demonstrates that the trypanolytic activity of Nbs and Fabs requires low molecular weight, monovalency and high affinity. We propose that the generation of low molecular weight VSG-specific trypanolytic nanobodies that impede endocytosis offers a new opportunity for developing novel trypanosomiasis therapeutics. In addition, these data suggest that the antigen-binding domain of an anti-microbial antibody harbours biological functionality that is latent in the intact immunoglobulin and is revealed only upon release of the antigen-binding fragment.


Asunto(s)
Anticuerpos Antiprotozoarios/farmacología , Endocitosis/efectos de los fármacos , Trypanosoma brucei brucei/inmunología , Glicoproteínas Variantes de Superficie de Trypanosoma/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/inmunología , Anticuerpos Antiprotozoarios/uso terapéutico , Afinidad de Anticuerpos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Nanopartículas , Tripanocidas/farmacología , Tripanocidas/uso terapéutico , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/fisiología , Trypanosoma brucei brucei/ultraestructura , Tripanosomiasis Africana/inmunología , Tripanosomiasis Africana/metabolismo , Tripanosomiasis Africana/terapia
16.
Nat Med ; 12(5): 580-4, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16604085

RESUMEN

High systemic drug toxicity and increasing prevalence of drug resistance hampers efficient treatment of human African trypanosomiasis (HAT). Hence, development of new highly specific trypanocidal drugs is necessary. Normal human serum (NHS) contains apolipoprotein L-I (apoL-I), which lyses African trypanosomes except resistant forms such as Trypanosoma brucei rhodesiense. T. b. rhodesiense expresses the apoL-I-neutralizing serum resistance-associated (SRA) protein, endowing this parasite with the ability to infect humans and cause HAT. A truncated apoL-I (Tr-apoL-I) has been engineered by deleting its SRA-interacting domain, which makes it lytic for T. b. rhodesiense. Here, we conjugated Tr-apoL-I with a single-domain antibody (nanobody) that efficiently targets conserved cryptic epitopes of the variant surface glycoprotein (VSG) of trypanosomes to generate a new manmade type of immunotoxin with potential for trypanosomiasis therapy. Treatment with this engineered conjugate resulted in clear curative and alleviating effects on acute and chronic infections of mice with both NHS-resistant and NHS-sensitive trypanosomes.


Asunto(s)
Apolipoproteínas/inmunología , Inmunotoxinas/uso terapéutico , Lipoproteínas HDL/inmunología , Tripanocidas/uso terapéutico , Tripanosomiasis Africana/tratamiento farmacológico , Glicoproteínas Variantes de Superficie de Trypanosoma/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Apolipoproteína L1 , Apolipoproteínas/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Lipoproteínas HDL/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Tripanocidas/inmunología , Trypanosoma brucei rhodesiense/inmunología , Trypanosoma brucei rhodesiense/metabolismo , Tripanosomiasis Africana/inmunología , Glicoproteínas Variantes de Superficie de Trypanosoma/genética
17.
Annu Rev Pathol ; 18: 19-45, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36055769

RESUMEN

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.


Asunto(s)
COVID-19 , Tripanosomiasis Africana , Humanos , Apolipoproteína L1/genética , Inflamación , Trypanosoma brucei rhodesiense/fisiología , Tripanosomiasis Africana/genética , Tripanosomiasis Africana/parasitología
18.
Cell Rep ; 42(12): 113528, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38041817

RESUMEN

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.


Asunto(s)
Apolipoproteína L1 , Membranas Mitocondriales , Apolipoproteína L1/genética , Membranas Mitocondriales/metabolismo , Aparato de Golgi/metabolismo , Mitocondrias , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Dinámicas Mitocondriales
19.
Trends Parasitol ; 38(2): 104-108, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34887168

RESUMEN

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.


Asunto(s)
Enfermedades Renales , Podocitos , Trypanosoma , Tripanosomiasis Africana , Animales , Apolipoproteína L1/genética , Apolipoproteína L1/metabolismo , Humanos , Enfermedades Renales/metabolismo , Podocitos/metabolismo , Trypanosoma brucei rhodesiense
20.
J Biol Chem ; 285(3): 1773-80, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19923211

RESUMEN

Sterol 14alpha-demethylase (14DM, the CYP51 family of cytochrome P450) is an essential enzyme in sterol biosynthesis in eukaryotes. It serves as a major drug target for fungal diseases and can potentially become a target for treatment of human infections with protozoa. Here we present 1.9 A resolution crystal structures of 14DM from the protozoan pathogen Trypanosoma brucei, ligand-free and complexed with a strong chemically selected inhibitor N-1-(2,4-dichlorophenyl)-2-(1H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadi-azol-2-yl)benzamide that we previously found to produce potent antiparasitic effects in Trypanosomatidae. This is the first structure of a eukaryotic microsomal 14DM that acts on sterol biosynthesis, and it differs profoundly from that of the water-soluble CYP51 family member from Mycobacterium tuberculosis, both in organization of the active site cavity and in the substrate access channel location. Inhibitor binding does not cause large scale conformational rearrangements, yet induces unanticipated local alterations in the active site, including formation of a hydrogen bond network that connects, via the inhibitor amide group fragment, two remote functionally essential protein segments and alters the heme environment. The inhibitor binding mode provides a possible explanation for both its functionally irreversible effect on the enzyme activity and its selectivity toward the 14DM from human pathogens versus the human 14DM ortholog. The structures shed new light on 14DM functional conservation and open an excellent opportunity for directed design of novel antiparasitic drugs.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450 , Trypanosoma brucei brucei/enzimología , Tripanosomiasis Africana/tratamiento farmacológico , Secuencia de Aminoácidos , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacología , Benzamidas/uso terapéutico , Biocatálisis , Cristalografía por Rayos X , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ligandos , Microsomas/enzimología , Modelos Moleculares , Conformación Proteica , Esterol 14-Desmetilasa , Esteroles/biosíntesis , Especificidad por Sustrato , Tripanocidas/química , Tripanocidas/metabolismo , Tripanocidas/farmacología , Tripanocidas/uso terapéutico , Trypanosoma brucei brucei/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA