Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
EMBO Rep ; 21(11): e50829, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33124769

RESUMEN

Inflammatory caspase-11 (rodent) and caspases-4/5 (humans) detect the Gram-negative bacterial component LPS within the host cell cytosol, promoting activation of the non-canonical inflammasome. Although non-canonical inflammasome-induced pyroptosis and IL-1-related cytokine release are crucial to mount an efficient immune response against various bacteria, their unrestrained activation drives sepsis. This suggests that cellular components tightly control the threshold level of the non-canonical inflammasome in order to ensure efficient but non-deleterious inflammatory responses. Here, we show that the IFN-inducible protein Irgm2 and the ATG8 family member Gate-16 cooperatively counteract Gram-negative bacteria-induced non-canonical inflammasome activation, both in cultured macrophages and in vivo. Specifically, the Irgm2/Gate-16 axis dampens caspase-11 targeting to intracellular bacteria, which lowers caspase-11-mediated pyroptosis and cytokine release. Deficiency in Irgm2 or Gate16 induces both guanylate binding protein (GBP)-dependent and GBP-independent routes for caspase-11 targeting to intracellular bacteria. Our findings identify molecular effectors that fine-tune bacteria-activated non-canonical inflammasome responses and shed light on the understanding of the immune pathways they control.


Asunto(s)
Caspasas , Lipopolisacáridos , Familia de las Proteínas 8 Relacionadas con la Autofagia , Caspasas/genética , Caspasas Iniciadoras , Bacterias Gramnegativas , Inflamasomas/genética , Macrófagos
2.
Proc Natl Acad Sci U S A ; 112(33): E4581-90, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26240314

RESUMEN

IFN-γ orchestrates cell-autonomous host defense against various intracellular vacuolar pathogens. IFN-γ-inducible GTPases, such as p47 immunity-related GTPases (IRGs) and p65 guanylate-binding proteins (GBPs), are recruited to pathogen-containing vacuoles, which is important for disruption of the vacuoles, culminating in the cell-autonomous clearance. Although the positive regulation for the proper recruitment of IRGs and GBPs to the vacuoles has been elucidated, the suppressive mechanism is unclear. Here, we show that Rab GDP dissociation inhibitor α (RabGDIα), originally identified as a Rab small GTPase inhibitor, is a negative regulator of IFN-γ-inducible GTPases in cell-autonomous immunity to the intracellular pathogen Toxoplasma gondii. Overexpression of RabGDIα, but not of RabGDIß, impaired IFN-γ-dependent reduction of T. gondii numbers. Conversely, RabGDIα deletion in macrophages and fibroblasts enhanced the IFN-γ-induced clearance of T. gondii. Furthermore, upon a high dose of infection by T. gondii, RabGDIα-deficient mice exhibited a decreased parasite burden in the brain and increased resistance in the chronic phase than did control mice. Among members of IRGs and GBPs important for the parasite clearance, Irga6 and Gbp2 alone were more frequently recruited to T. gondii-forming parasitophorous vacuoles in RabGDIα-deficient cells. Notably, Gbp2 positively controlled Irga6 recruitment that was inhibited by direct and specific interactions of RabGDIα with Gbp2 through the lipid-binding pocket. Taken together, our results suggest that RabGDIα inhibits host defense against T. gondii by negatively regulating the Gbp2-Irga6 axis of IFN-γ-dependent cell-autonomous immunity.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Regulación Enzimológica de la Expresión Génica , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Interferón gamma/inmunología , Toxoplasma/patogenicidad , Toxoplasmosis/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Chlorocebus aethiops , Cartilla de ADN/genética , Femenino , Fibroblastos/metabolismo , Inflamación/inmunología , Lípidos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido , Células Vero
3.
Cell Microbiol ; 18(2): 244-59, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26247512

RESUMEN

In mice, avirulent strains (e.g. types II and III) of the protozoan parasite Toxoplasma gondii are restricted by the immunity-related GTPase (IRG) resistance system. Loading of IRG proteins onto the parasitophorous vacuolar membrane (PVM) is required for vacuolar rupture resulting in parasite clearance. In virulent strain (e.g. type I) infections, polymorphic effector proteins ROP5 and ROP18 cooperate to phosphorylate and thereby inactivate mouse IRG proteins to preserve PVM integrity. In this study, we confirmed the dense granule protein GRA7 as an additional component of the ROP5/ROP18 kinase complex and identified GRA7 association with the PVM by direct binding to ROP5. The absence of GRA7 results in reduced phosphorylation of Irga6 correlated with increased vacuolar IRG protein amounts and attenuated virulence. Earlier work identified additional IRG proteins as targets of T. gondii ROP18 kinase. We show that the only specific target of ROP18 among IRG proteins is in fact Irga6. Similarly, we demonstrate that GRA7 is strictly an Irga6-specific virulence effector. This identifies T. gondii GRA7 as a regulator for ROP18-specific inactivation of Irga6. The structural diversity of the IRG proteins implies that certain family members constitute additional specific targets for other yet unknown T. gondii virulence effectors.


Asunto(s)
Antígenos de Protozoos/metabolismo , GTP Fosfohidrolasas/metabolismo , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/genética , Toxoplasma/fisiología , Animales , Ratones , Unión Proteica
4.
BMC Biol ; 14: 33, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27098192

RESUMEN

BACKGROUND: The interferon-γ (IFN-γ)-inducible immunity-related GTPase (IRG), Irgm1, plays an essential role in restraining activation of the IRG pathogen resistance system. However, the loss of Irgm1 in mice also causes a dramatic but unexplained susceptibility phenotype upon infection with a variety of pathogens, including many not normally controlled by the IRG system. This phenotype is associated with lymphopenia, hemopoietic collapse, and death of the mouse. RESULTS: We show that the three regulatory IRG proteins (GMS sub-family), including Irgm1, each of which localizes to distinct sets of endocellular membranes, play an important role during the cellular response to IFN-γ, each protecting specific membranes from off-target activation of effector IRG proteins (GKS sub-family). In the absence of Irgm1, which is localized mainly at lysosomal and Golgi membranes, activated GKS proteins load onto lysosomes, and are associated with reduced lysosomal acidity and failure to process autophagosomes. Another GMS protein, Irgm3, is localized to endoplasmic reticulum (ER) membranes; in the Irgm3-deficient mouse, activated GKS proteins are found at the ER. The Irgm3-deficient mouse does not show the drastic phenotype of the Irgm1 mouse. In the Irgm1/Irgm3 double knock-out mouse, activated GKS proteins associate with lipid droplets, but not with lysosomes, and the Irgm1/Irgm3(-/-) does not have the generalized immunodeficiency phenotype expected from its Irgm1 deficiency. CONCLUSIONS: The membrane targeting properties of the three GMS proteins to specific endocellular membranes prevent accumulation of activated GKS protein effectors on the corresponding membranes and thus enable GKS proteins to distinguish organellar cellular membranes from the membranes of pathogen vacuoles. Our data suggest that the generalized lymphomyeloid collapse that occurs in Irgm1(-/-) mice upon infection with a variety of pathogens may be due to lysosomal damage caused by off-target activation of GKS proteins on lysosomal membranes and consequent failure of autophagosomal processing.


Asunto(s)
GTP Fosfohidrolasas/inmunología , Proteínas de Unión al GTP/inmunología , Lisosomas/inmunología , Animales , Autofagia , Línea Celular , GTP Fosfohidrolasas/análisis , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/análisis , Proteínas de Unión al GTP/genética , Eliminación de Gen , Inmunidad Innata , Infecciones/genética , Infecciones/inmunología , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
PLoS Pathog ; 10(10): e1004449, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25356593

RESUMEN

The IRG system of IFNγ-inducible GTPases constitutes a powerful resistance mechanism in mice against Toxoplasma gondii and two Chlamydia strains but not against many other bacteria and protozoa. Why only T. gondii and Chlamydia? We hypothesized that unusual features of the entry mechanisms and intracellular replicative niches of these two organisms, neither of which resembles a phagosome, might hint at a common principle. We examined another unicellular parasitic organism of mammals, member of an early-diverging group of Fungi, that bypasses the phagocytic mechanism when it enters the host cell: the microsporidian Encephalitozoon cuniculi. Consistent with the known susceptibility of IFNγ-deficient mice to E. cuniculi infection, we found that IFNγ treatment suppresses meront development and spore formation in mouse fibroblasts in vitro, and that this effect is mediated by IRG proteins. The process resembles that previously described in T. gondii and Chlamydia resistance. Effector (GKS subfamily) IRG proteins accumulate at the parasitophorous vacuole of E. cuniculi and the meronts are eliminated. The suppression of E. cuniculi growth by IFNγ is completely reversed in cells lacking regulatory (GMS subfamily) IRG proteins, cells that effectively lack all IRG function. In addition IFNγ-induced cells infected with E. cuniculi die by necrosis as previously shown for IFNγ-induced cells resisting T. gondii infection. Thus the IRG resistance system provides cell-autonomous immunity to specific parasites from three kingdoms of life: protozoa, bacteria and fungi. The phylogenetic divergence of the three organisms whose vacuoles are now known to be involved in IRG-mediated immunity and the non-phagosomal character of the vacuoles themselves strongly suggests that the IRG system is triggered not by the presence of specific parasite components but rather by absence of specific host components on the vacuolar membrane.


Asunto(s)
Encephalitozoon cuniculi/inmunología , Encefalitozoonosis/inmunología , GTP Fosfohidrolasas/inmunología , Interferón gamma/inmunología , Animales , Supervivencia Celular , Encephalitozoon cuniculi/crecimiento & desarrollo , Encefalitozoonosis/microbiología , Fibroblastos , GTP Fosfohidrolasas/biosíntesis , GTP Fosfohidrolasas/genética , Inmunidad Innata , Membranas Intracelulares/inmunología , Ratones , Ratones Endogámicos C57BL , Necrosis , Fagosomas/inmunología , Vacuolas/inmunología
6.
PLoS Biol ; 10(7): e1001358, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22802726

RESUMEN

The ability of mice to resist infection with the protozoan parasite, Toxoplasma gondii, depends in large part on the function of members of a complex family of atypical large GTPases, the interferon-gamma-inducible immunity-related GTPases (IRG proteins). Nevertheless, some strains of T. gondii are highly virulent for mice because, as recently shown, they secrete a polymorphic protein kinase, ROP18, from the rhoptries into the host cell cytosol at the moment of cell invasion. Depending on the allele, ROP18 can act as a virulence factor for T. gondii by phosphorylating and thereby inactivating mouse IRG proteins. In this article we show that IRG proteins interact not only with ROP18, but also strongly with the products of another polymorphic locus, ROP5, already implicated as a major virulence factor from genetic crosses, but whose function has previously been a complete mystery. ROP5 proteins are members of the same protein family as ROP18 kinases but are pseudokinases by sequence, structure, and function. We show by a combination of genetic and biochemical approaches that ROP5 proteins act as essential co-factors for ROP18 and present evidence that they work by enforcing an inactive GDP-dependent conformation on the IRG target protein. By doing so they prevent GTP-dependent activation and simultaneously expose the target threonines on the switch I loop for phosphorylation by ROP18, resulting in permanent inactivation of the protein. This represents a novel mechanism in which a pseudokinase facilitates the phosphorylation of a target by a partner kinase by preparing the substrate for phosphorylation, rather than by upregulation of the activity of the kinase itself.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Toxoplasma/enzimología , Animales , Dominio Catalítico , Células Cultivadas , Citosol/metabolismo , Activación Enzimática , Fibroblastos/parasitología , GTP Fosfohidrolasas/genética , Interacciones Huésped-Parásitos , Inmunohistoquímica , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Organismos Modificados Genéticamente/genética , Organismos Modificados Genéticamente/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Protozoarias , Treonina/genética , Treonina/metabolismo , Toxoplasma/genética , Toxoplasma/patogenicidad , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
7.
J Immunol ; 191(4): 1765-74, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23842753

RESUMEN

The IFN-inducible protein Irgm1 (LRG-47) belongs to the family of immunity-related GTPases that function in cell-autonomous resistance against intracellular pathogens in mice. Irgm1 deficiency is associated with a severe immunodeficiency syndrome. The protein has been variously interpreted as a direct effector molecule on bacterial phagosomes or on other organelles or as an inducer of autophagy. In this study, we re-examined one of these claims, namely that Irgm1 targets mycobacterial and listerial phagosomes. We found no colocalization of endogenous Irgm1, using two immunofluorescent staining techniques, either in fibroblasts or in macrophages. We demonstrated the predicted existence of two protein isoforms of Irgm1 derived from differential splicing and described immunological reagents for their detection. Both Irgm1 isoforms localize to the Golgi apparatus and weakly to mitochondria; however, only the long Irgm1 isoforms can be detected on endolysosomal membranes. Together with the previous observation that the general immunodeficiency phenotype of Irgm1(-/-) mice is reversed in Irgm1/Irgm3 double-deficient mice, our results argue against a direct effector function of Irgm1 at the bacterial phagosome. We discuss these findings in the context of evidence that Irgm1 functions as a negative regulator of other members of the immunity-related GTPase protein family.


Asunto(s)
Proteínas de Unión al GTP/inmunología , Interferón gamma/farmacología , Fagosomas/inmunología , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Línea Celular , Fibroblastos/química , Técnica del Anticuerpo Fluorescente Directa , GTP Fosfohidrolasas/clasificación , Proteínas de Unión al GTP/análisis , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Aparato de Golgi/química , Humanos , Inmunización , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Listeria monocytogenes/inmunología , Macrófagos/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/química , Datos de Secuencia Molecular , Mycobacterium bovis/inmunología , Fragmentos de Péptidos/inmunología , Fagosomas/microbiología , Isoformas de Proteínas/análisis , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología
8.
PLoS Pathog ; 8(3): e1002567, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457617

RESUMEN

Toxoplasma gondii is a zoonotic protozoan parasite which infects nearly one third of the human population and is found in an extraordinary range of vertebrate hosts. Its epidemiology depends heavily on horizontal transmission, especially between rodents and its definitive host, the cat. Neospora caninum is a recently discovered close relative of Toxoplasma, whose definitive host is the dog. Both species are tissue-dwelling Coccidia and members of the phylum Apicomplexa; they share many common features, but Neospora neither infects humans nor shares the same wide host range as Toxoplasma, rather it shows a striking preference for highly efficient vertical transmission in cattle. These species therefore provide a remarkable opportunity to investigate mechanisms of host restriction, transmission strategies, virulence and zoonotic potential. We sequenced the genome of N. caninum and transcriptomes of the invasive stage of both species, undertaking an extensive comparative genomics and transcriptomics analysis. We estimate that these organisms diverged from their common ancestor around 28 million years ago and find that both genomes and gene expression are remarkably conserved. However, in N. caninum we identified an unexpected expansion of surface antigen gene families and the divergence of secreted virulence factors, including rhoptry kinases. Specifically we show that the rhoptry kinase ROP18 is pseudogenised in N. caninum and that, as a possible consequence, Neospora is unable to phosphorylate host immunity-related GTPases, as Toxoplasma does. This defense strategy is thought to be key to virulence in Toxoplasma. We conclude that the ecological niches occupied by these species are influenced by a relatively small number of gene products which operate at the host-parasite interface and that the dominance of vertical transmission in N. caninum may be associated with the evolution of reduced virulence in this species.


Asunto(s)
Coccidiosis/parasitología , Genómica , Neospora/genética , Toxoplasma/genética , Toxoplasmosis/parasitología , Animales , Coccidiosis/transmisión , Hibridación Genómica Comparativa , Regulación de la Expresión Génica , Interacciones Huésped-Parásitos/fisiología , Transmisión Vertical de Enfermedad Infecciosa , Neospora/patogenicidad , Toxoplasma/patogenicidad , Toxoplasmosis/transmisión , Virulencia , Zoonosis/transmisión
9.
Cell Microbiol ; 14(12): 1921-33, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22906355

RESUMEN

Toxoplasma gondii uses specialized secretory organelles called rhoptries to deliver virulence determinants into the host cell during parasite invasion. One such determinant called rhoptry protein 18 (ROP18) is a polymorphic serine/threonine kinase that phosphorylates host targets to modulate acute virulence. Following secretion into the host cell, ROP18 traffics to the parasitophorous vacuole membrane (PVM) where it is tethered to the cytosolic face of this host-pathogen interface. However, the functional consequences of PVM association are not known. In this report, we show that ROP18 mutants altered in an arginine-rich domain upstream of the kinase domain fail to associate to the PVM following secretion from rhoptries. During infection, host cells upregulate immunity-related GTPases that localize to and destroy the PVM surrounding the parasites. ROP18 disarms this host innate immune pathway by phosphorylating IRGs in a critical GTPase domain and preventing loading on the PVM. Vacuole-targeting mutants of ROP18 failed to phosphorylate Irga6 and were unable to divert IRGs from the PVM, despite retaining intrinsic kinase activity. As a consequence, these mutants were avirulent in a mouse model of acute toxoplasmosis. Thus, the association of ROP18 with the PVM, mediated by its N-terminal arginine-rich domain, is critical to its function as a virulence determinant.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Toxoplasma/patogenicidad , Vacuolas/metabolismo , Vacuolas/parasitología , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Protozoarias , Eliminación de Secuencia , Toxoplasmosis Animal/parasitología , Toxoplasmosis Animal/patología , Virulencia , Factores de Virulencia/deficiencia , Factores de Virulencia/genética
11.
PLoS Biol ; 8(12): e1000576, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21203588

RESUMEN

Virulence of complex pathogens in mammals is generally determined by multiple components of the pathogen interacting with the functional complexity and multiple layering of the mammalian immune system. It is most unusual for the resistance of a mammalian host to be overcome by the defeat of a single defence mechanism. In this study we uncover and analyse just such a case at the molecular level, involving the widespread intracellular protozoan pathogen Toxoplasma gondii and one of its most important natural hosts, the house mouse (Mus musculus). Natural polymorphism in virulence of Eurasian T. gondii strains for mice has been correlated in genetic screens with the expression of polymorphic rhoptry kinases (ROP kinases) secreted into the host cell during infection. We show that the molecular targets of the virulent allelic form of ROP18 kinase are members of a family of cellular GTPases, the interferon-inducible IRG (immunity-related GTPase) proteins, known from earlier work to be essential resistance factors in mice against avirulent strains of T. gondii. Virulent T. gondii strain ROP18 kinase phosphorylates several mouse IRG proteins. We show that the parasite kinase phosphorylates host Irga6 at two threonines in the nucleotide-binding domain, biochemically inactivating the GTPase and inhibiting its accumulation and action at the T. gondii parasitophorous vacuole membrane. Our analysis identifies the conformationally active switch I region of the GTP-binding site as an Achilles' heel of the IRG protein pathogen-resistance mechanism. The polymorphism of ROP18 in natural T. gondii populations indicates the existence of a dynamic, rapidly evolving ecological relationship between parasite virulence factors and host resistance factors. This system should be unusually fruitful for analysis at both ecological and molecular levels since both T. gondii and the mouse are widespread and abundant in the wild and are well-established model species with excellent analytical tools available.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas/inmunología , Toxoplasma/inmunología , Toxoplasma/patogenicidad , Animales , Membrana Celular/química , Membrana Celular/parasitología , Membrana Celular/fisiología , Permeabilidad de la Membrana Celular , Interacciones Huésped-Parásitos , Evasión Inmune , Proteínas de la Membrana/metabolismo , Ratones , Fosforilación , Polimorfismo Genético , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Protozoarias/metabolismo , Toxoplasma/fisiología , Vacuolas/química , Vacuolas/parasitología , Vacuolas/fisiología , Virulencia
12.
J Biol Chem ; 286(35): 30471-30480, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21757726

RESUMEN

The immunity-related GTPases (IRGs) are a family of proteins induced by interferon-γ that play a crucial role in innate resistance to intracellular pathogens. The M subfamily of IRG proteins (IRGM) plays a profound role in this context, in part because of the ability of its members to regulate the localization and expression of other IRG proteins. We present here evidence that IRGM proteins affect the localization of the guanylate-binding proteins (GBPs), a second family of interferon-induced GTP-binding proteins that also function in innate immunity. Absence of Irgm1 or Irgm3 led to accumulation of Gbp2 in intracellular compartments that were positive for both the macroautophagy (hereafter referred to as autophagy) marker LC3 and the autophagic adapter molecule p62/Sqstm1. Gbp2 was similarly relocalized in cells in which autophagy was impaired because of the absence of Atg5. Both in Atg5- and IRGM-deficient cells, the IRG protein Irga6 relocalized to the same compartments as Gbp2, raising the possibility of a common regulatory mechanism. However, other data indicated that Irga6, but not Gbp2, was ubiquitinated in IRGM-deficient cells. Similarly, coimmunoprecipitation studies indicated that although Irgm3 did interact directly with Irgb6, it did not interact with Gbp2. Collectively, these data suggest that IRGM proteins indirectly modulate the localization of GBPs through a distinct mechanism from that through which they regulate IRG protein localization. Further, these results suggest that a core function of IRGM proteins is to regulate autophagic flux, which influences the localization of GBPs and possibly other factors that instruct cell-autonomous immune resistance.


Asunto(s)
Autofagia , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Células 3T3 , Animales , Flavonoles , Glicósidos , Inmunoprecipitación , Interferones/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Fagosomas/metabolismo , Unión Proteica , Ubiquitina/metabolismo
13.
EMBO J ; 27(19): 2495-509, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-18772884

RESUMEN

Members of the immunity-related GTPase (IRG) family are interferon-inducible resistance factors against a broad spectrum of intracellular pathogens including Toxoplasma gondii. The molecular mechanisms governing the function and regulation of the IRG resistance system are largely unknown. We find that IRG proteins function in a system of direct, nucleotide-dependent regulatory interactions between family members. After interferon induction but before infection, the three members of the GMS subfamily of IRG proteins, Irgm1, Irgm2 and Irgm3, which possess an atypical nucleotide-binding site, regulate the intracellular positioning of the conventional GKS subfamily members, Irga6 and Irgb6. Following infection, the normal accumulation of Irga6 protein at the parasitophorous vacuole membrane (PVM) is nucleotide dependent and also depends on the presence of all three GMS proteins. We present evidence that an essential role of the GMS proteins in this response is control of the nucleotide-bound state of the GKS proteins, preventing their GTP-dependent activation before infection. Accumulation of IRG proteins at the PVM has previously been shown to be associated with a block in pathogen replication: our results relate for the first time the enzymatic properties of IRG proteins to their role in pathogen resistance.


Asunto(s)
Proteínas de Unión al GTP/inmunología , Inmunidad Innata/fisiología , Interferones/inmunología , Toxoplasma/inmunología , Animales , Línea Celular , Fibroblastos/citología , Fibroblastos/microbiología , Fibroblastos/fisiología , Proteínas de Unión al GTP/genética , Ratones , Toxoplasma/patogenicidad , Técnicas del Sistema de Dos Híbridos
14.
PLoS Pathog ; 6(8): e1001071, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20865117

RESUMEN

UNC93B1 associates with Toll-Like Receptor (TLR) 3, TLR7 and TLR9, mediating their translocation from the endoplasmic reticulum to the endolysosome, hence allowing proper activation by nucleic acid ligands. We found that the triple deficient '3d' mice, which lack functional UNC93B1, are hyper-susceptible to infection with Toxoplasma gondii. We established that while mounting a normal systemic pro-inflammatory response, i.e. producing abundant MCP-1, IL-6, TNFα and IFNγ, the 3d mice were unable to control parasite replication. Nevertheless, infection of reciprocal bone marrow chimeras between wild-type and 3d mice with T. gondii demonstrated a primary role of hemopoietic cell lineages in the enhanced susceptibility of UNC93B1 mutant mice. The protective role mediated by UNC93B1 to T. gondii infection was associated with impaired IL-12 responses and delayed IFNγ by spleen cells. Notably, in macrophages infected with T. gondii, UNC93B1 accumulates on the parasitophorous vacuole. Furthermore, upon in vitro infection the rate of tachyzoite replication was enhanced in non-activated macrophages carrying mutant UNC93B1 as compared to wild type gene. Strikingly, the role of UNC93B1 on intracellular parasite growth appears to be independent of TLR function. Altogether, our results reveal a critical role for UNC93B1 on induction of IL-12/IFNγ production as well as autonomous control of Toxoplasma replication by macrophages.


Asunto(s)
Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/inmunología , Toxoplasmosis Animal/genética , Toxoplasmosis Animal/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Immunoblotting , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Confocal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Toxoplasma/genética , Toxoplasma/inmunología
15.
PLoS Genet ; 5(3): e1000403, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19266026

RESUMEN

Immunity-related GTPases (IRG) play an important role in defense against intracellular pathogens. One member of this gene family in humans, IRGM, has been recently implicated as a risk factor for Crohn's disease. We analyzed the detailed structure of this gene family among primates and showed that most of the IRG gene cluster was deleted early in primate evolution, after the divergence of the anthropoids from prosimians ( about 50 million years ago). Comparative sequence analysis of New World and Old World monkey species shows that the single-copy IRGM gene became pseudogenized as a result of an Alu retrotransposition event in the anthropoid common ancestor that disrupted the open reading frame (ORF). We find that the ORF was reestablished as a part of a polymorphic stop codon in the common ancestor of humans and great apes. Expression analysis suggests that this change occurred in conjunction with the insertion of an endogenous retrovirus, which altered the transcription initiation, splicing, and expression profile of IRGM. These data argue that the gene became pseudogenized and was then resurrected through a series of complex structural events and suggest remarkable functional plasticity where alleles experience diverse evolutionary pressures over time. Such dynamism in structure and evolution may be critical for a gene family locked in an arms race with an ever-changing repertoire of intracellular parasites.


Asunto(s)
Evolución Molecular , Proteínas de Unión al GTP/genética , Animales , Expresión Génica , Humanos , Familia de Multigenes , Mutagénesis Insercional , Filogenia , Primates/clasificación , Primates/genética , Seudogenes , Retroelementos
16.
BMC Biol ; 9: 7, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21276251

RESUMEN

BACKGROUND: The interferon-inducible immunity-related GTPases (IRG proteins/p47 GTPases) are a distinctive family of GTPases that function as powerful cell-autonomous resistance factors. The IRG protein, Irga6 (IIGP1), participates in the disruption of the vacuolar membrane surrounding the intracellular parasite, Toxoplasma gondii, through which it communicates with its cellular hosts. Some aspects of the protein's behaviour have suggested a dynamin-like molecular mode of action, in that the energy released by GTP hydrolysis is transduced into mechanical work that results in deformation and ultimately rupture of the vacuolar membrane. RESULTS: Irga6 forms GTP-dependent oligomers in vitro and thereby activates hydrolysis of the GTP substrate. In this study we define the catalytic G-domain interface by mutagenesis and present a structural model, of how GTP hydrolysis is activated in Irga6 complexes, based on the substrate-twinning reaction mechanism of the signal recognition particle (SRP) and its receptor (SRα). In conformity with this model, we show that the bound nucleotide is part of the catalytic interface and that the 3'hydroxyl of the GTP ribose bound to each subunit is essential for trans-activation of hydrolysis of the GTP bound to the other subunit. We show that both positive and negative regulatory interactions between IRG proteins occur via the catalytic interface. Furthermore, mutations that disrupt the catalytic interface also prevent Irga6 from accumulating on the parasitophorous vacuole membrane of T. gondii, showing that GTP-dependent Irga6 activation is an essential component of the resistance mechanism. CONCLUSIONS: The catalytic interface of Irga6 defined in the present experiments can probably be used as a paradigm for the nucleotide-dependent interactions of all members of the large family of IRG GTPases, both activating and regulatory. Understanding the activation mechanism of Irga6 will help to explain the mechanism by which IRG proteins exercise their resistance function. We find no support from sequence or G-domain structure for the idea that IRG proteins and the SRP GTPases have a common phylogenetic origin. It therefore seems probable, if surprising, that the substrate-assisted catalytic mechanism has been independently evolved in the two protein families.


Asunto(s)
GTP Fosfohidrolasas/inmunología , Inmunidad Innata , Toxoplasma/metabolismo , Vacuolas/inmunología , Animales , GTP Fosfohidrolasas/metabolismo , Guanosina Trifosfato/metabolismo , Interacciones Huésped-Parásitos , Ratones , Ratones Endogámicos C57BL , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Péptidos/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Vacuolas/metabolismo , Vacuolas/parasitología
17.
Mamm Genome ; 22(1-2): 43-54, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21052678

RESUMEN

The immunity-related GTPases (IRGs) belong to the family of large, interferon-inducible GTPases and constitute a cell-autonomous resistance system essential for the control of vacuolar pathogens like Toxoplasma gondii in mice. Recent results demonstrated that numerous IRG members accumulate collaboratively at the parasitophorous vacuole of invading T. gondii leading to the destruction of the vacuole and the parasite and subsequent necrotic host cell death. Complex regulatory interactions between different IRG proteins are necessary for these processes. Disturbance of this finely balanced system, e.g., by single genetic deficiency for the important negative regulator Irgm1 or the autophagic regulator Atg5, leads to spontaneous activation of the effector IRG proteins when induced by IFNγ. This activation has cytotoxic consequences resulting in a severe lymphopenia, macrophage defects, and failure of the adaptive immune system in Irgm1-deficient mice. However, alternative functions in phagosome maturation and induction of autophagy have been proposed for Irgm1. The IRG system has been studied primarily in mice, but IRG genes are present throughout the mammalian lineage. Interestingly, the number, type, and diversity of genes present differ greatly even between closely related species, probably reflecting intimate host-pathogen coevolution driven by an armed race between the IRG resistance proteins and pathogen virulence factors. IRG proteins are targets for polymorphic T. gondii virulence factors, and genetic variation in the IRG system between different mouse strains correlates with resistance and susceptibility to virulent T. gondii strains.


Asunto(s)
GTP Fosfohidrolasas/inmunología , Interacciones Huésped-Patógeno , Mamíferos/inmunología , Animales , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/genética , Regulación de la Expresión Génica , Humanos , Mamíferos/microbiología , Mamíferos/parasitología , Mamíferos/virología , Ratones , Fagosomas/inmunología
18.
PLoS Pathog ; 5(2): e1000288, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19197351

RESUMEN

Toxoplasma gondii is a natural intracellular protozoal pathogen of mice and other small mammals. After infection, the parasite replicates freely in many cell types (tachyzoite stage) before undergoing a phase transition and encysting in brain and muscle (bradyzoite stage). In the mouse, early immune resistance to the tachyzoite stage is mediated by the family of interferon-inducible immunity-related GTPases (IRG proteins), but little is known of the nature of this resistance. We reported earlier that IRG proteins accumulate on intracellular vacuoles containing the pathogen, and that the vacuolar membrane subsequently ruptures. In this report, live-cell imaging microscopy has been used to follow this process and its consequences in real time. We show that the rupture of the vacuole is inevitably followed by death of the intracellular parasite, shown by its permeability to cytosolic protein markers. Death of the parasite is followed by the death of the infected cell. The death of the cell has features of pyronecrosis, including membrane permeabilisation and release of the inflammatory protein, HMGB1, but caspase-1 cleavage is not detected. This sequence of events occurs on a large scale only following infection of IFNgamma-induced cells with an avirulent strain of T. gondii, and is reduced by expression of a dominant negative mutant IRG protein. Cells infected by virulent strains rarely undergo necrosis. We did not find autophagy to play any role in the key steps leading to the death of the parasite. We conclude that IRG proteins resist infection by avirulent T. gondii by a novel mechanism involving disruption of the vacuolar membrane, which in turn ultimately leads to the necrotic death of the infected cell.


Asunto(s)
GTP Fosfohidrolasas/inmunología , Interferón gamma/inmunología , Necrosis/inmunología , Toxoplasma/metabolismo , Toxoplasmosis Animal/parasitología , Vacuolas/inmunología , Animales , Autofagia/inmunología , Fibroblastos/inmunología , Fibroblastos/parasitología , GTP Fosfohidrolasas/genética , Ratones , Microscopía Fluorescente , Microscopía de Contraste de Fase , Necrosis/parasitología , Toxoplasma/genética , Vacuolas/parasitología
19.
Cell Microbiol ; 12(7): 939-61, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20109161

RESUMEN

The immunity-related GTPases (IRGs) constitute an interferon-induced intracellular resistance mechanism in mice against Toxoplasma gondii. IRG proteins accumulate on the parasitophorous vacuole membrane (PVM), leading to its disruption and to death of the parasite. How IRGs target the PVM is unknown. We show that accumulation of IRGs on the PVM begins minutes after parasite invasion and increases for about 1 h. Targeting occurs independently of several signalling pathways and the microtubule network, suggesting that IRG transport is diffusion-driven. The intensity of IRG accumulation on the PVM, however, is reduced in absence of the autophagy regulator, Atg5. In wild-type cells IRG proteins accumulate cooperatively on PVMs in a definite order reflecting a temporal hierarchy, with Irgb6 and Irgb10 apparently acting as pioneers. Loading of IRG proteins onto the vacuoles of virulent Toxoplasma strains is attenuated and the two pioneer IRGs are the most affected. The polymorphic rhoptry kinases, ROP16, ROP18 and the catalytically inactive proteins, ROP5A-D, are not individually responsible for this effect. Thus IRG proteins protect mice against avirulent strains of Toxoplasma but fail against virulent strains. The complex cooperative behaviour of IRG proteins in resisting Toxoplasma may hint at undiscovered complexity also in virulence mechanisms.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Toxoplasma/inmunología , Vacuolas/enzimología , Vacuolas/parasitología , Animales , Western Blotting , Línea Celular , Electroforesis en Gel de Poliacrilamida , Inmunohistoquímica , Ratones
20.
J Immunol ; 182(6): 3775-81, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265156

RESUMEN

Cytokine-activated macrophages restrain the replication of intracellular parasites and disrupt the integrity of vacuolar pathogens. In this study, we show that inducible nitric oxide synthase and the immunity-related GTPase (IRG) family member Irgm3, respectively, are required for the ability of in vivo primed macrophages to restrain the growth of Toxoplasma gondii and to destroy the parasite's intracellular niche. Remarkably, virulent Type I strains of T. gondii evade IRG-dependent vacuolar disruption, while remaining susceptible to iNOS-dependent restriction. The ability of virulent T. gondii to escape killing by macrophages is controlled at the level of the individual vacuole and is associated with differential permissiveness for association of the IRG proteins Irga6 (IIGP1) and Irgb6 (TGTP) to the vacuolar membrane. Surprisingly, expression of the Type I ROP-18 virulence determinant in an avirulent strain did not confer the evasive phenotype. These results pinpoint evasion of vacuolar disruption by IRG proteins as a new determinant of pathogen virulence.


Asunto(s)
GTP Fosfohidrolasas/fisiología , Proteínas de Unión al GTP/fisiología , Activación de Macrófagos/inmunología , Toxoplasma/inmunología , Toxoplasma/patogenicidad , Vacuolas/enzimología , Vacuolas/inmunología , Animales , GTP Fosfohidrolasas/deficiencia , GTP Fosfohidrolasas/genética , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Toxoplasma/crecimiento & desarrollo , Vacuolas/parasitología , Virulencia/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA