IRGs

Last updated

Immunity Related Guanosine Triphosphatases or IRGs are proteins activated as part of an early immune response. IRGs have been described in various mammals but are most well characterized in mice. IRG activation in most cases is induced by an immune response and leads to clearance of certain pathogens.

Contents

Figure 1: Crystal structure of a Mouse IRG (PDB id 1TQ6) Mouse IRG.png
Figure 1: Crystal structure of a Mouse IRG (PDB id 1TQ6)

Background

Interferon (IFN)-inducible GTPases encompass four families of proteins including myxovirus resistant proteins (Mx), guanylate-binding proteins (GBP), immunity-related GTPase proteins (IRGs), and very large inducible GTPase proteins (VLIG). IRGs confer resistance from vacuolar pathogens by localizing to and disrupting the phagocytic vacuole during infection. The activation of IRGs in mice is induced by interferon. IRG genes have been identified in various vertebrates and some invertebrates. They are involved in important immune defenses against intracellular pathogens and as a result have become a target for immune evasion by those pathogens. The intracellular protozoan parasite Toxoplasma gondii has been shown to target IRGs in mice allowing for resistance from the host immune response.

Evolution of IRGs

IRGs Have Evolved From Invertebrates

Studies to determine the evolutionary origins of vertebrates have led to understanding the development of immune system processes and furthermore answer the questions of how and why pathogens have learned to evade and shut down these selectable genetic traits. Eight functional and four pseudo IRG genes have been identified in the invertebrate Branchiostoma floridiae . [1] Li et al. determined expression patterns of functional IFN-inducible GTPase genes in Branchiostoma japonicum at various immunologic sites when induced by pathogens and pathogenic substances. This evidence suggests that IRGs may function in an immune-related capacity in cephalochordates. The paradox remains that these IRGs function without induction by IFN activation pathways, since B. japonicum and other amphioxus species do not possess IFN and IFN receptor genes. [2] It is possible that IRGs may have existed prior to the Cambrian Explosion as an innate immune mechanism and with the evolution of the adaptive immune system in vertebrates, IFN evolved to modulate IRG function.

Vertebrates have evolved an array of IRG genes as a whole, potentially due to the evolution between variable pathogen interactions. The C57BL/6 mouse has 23 IRG genes of which 21 may be functional in resistance to pathogens (6 are well characterized), [3] whereas humans have evolved only 1 functional IRG gene (IRGM) and one pseudogene. [4] Studies in mice have characterized the importance of the type 2 effector molecule IFNγ in various cell types [5] [6] and gone on to determine the importance of these proteins in intracellular pathogen resistance. [7]

Orthologous Irgc (aka: Cinema) genes are found in humans and mice. These orthologs are not IFN inducible and are expressed only in the testis of both mammals. [3] Multiple IRG genes have been identified in canines and zebrafish but few in the model organism Tetraodontidae (the pufferfish). IRG genes in humans are thought to have been lost in the divergence of primates. [1] [3] The variations between and within species suggest a high rate of evolutionary change for this particular element of host pathogen interaction and highlight the importance of understanding the limitations of using model systems to study human immunology.

Mechanisms

Dependency on the IRGs is best exemplified in mouse studies. Multiple studies have been done using mouse knockout models to determine IRG function. Pathogen clearance mechanisms via lysosome maturation and vacuole destruction have been determined. Additionally, IRGs are implicated in the control of hematopoietic balance during infection. Irg1 knockout mice infected with Mycobacterium resulted in pancytopenia as a result of inadequate hematopoietic stem cell expansion. [8]

IRGs and Mice

Figure 2: Effects of Genetic Polymorphisms. Host polymorphisms within the IRG loci can alter the ability of cells to eliminate the vacuolated pathogen. Alternatively pathogens containing coding variations in effector proteins can circumvent the cellular response to infection to generate a pathogen permissive environment. Genetic Background Causation.jpg
Figure 2: Effects of Genetic Polymorphisms. Host polymorphisms within the IRG loci can alter the ability of cells to eliminate the vacuolated pathogen. Alternatively pathogens containing coding variations in effector proteins can circumvent the cellular response to infection to generate a pathogen permissive environment.

The mouse genome encodes 23 IRGs, several of which have been demonstrated to be widely expressed (liver, heart, spleen, intestine, thymus, lung, testis, kidney, brain, skin) in a number of cell types, [9] and are greatly up-regulated following exposure to the potent immune effector molecule interferon gamma, IFNγ. [10] IRGs are subdivided into two additional classes based on the mode of activity and mechanism. The GSK class (Irga6, Irgb6, and Irgd) are considered the canonical grouping of GTPases, whereas a second grouping of GMS proteins, which have a lysine to methionine mutation in the active site, function to prevent premature activation by associating with the nucleotide binding motif in a manner similar to Guanosine nucleotide dissociation inhibitors (GDI's). [11] [12] The subcellular localization of IRGs are variable; Irga6 and Irgm3 are predominantly found within the endoplasmic reticulum, Irgm1 and Irgm2 have been localized to the Golgi apparatus, [13] and at least two IRGs (Irgb6 and Irgd) have been found predominantly within the cytosol. [14] Following cellular entry of Toxoplasma gondii, IRGs can quickly redistribute onto the parasitophorous vacuole membrane (PVM) within 2–30 minutes. [12] The approximate order of decorating the PVM has been defined starting with the loading of Irgb6 and Irgb10 followed by Irga6, Irgd and Irgm2. Faint localization of Irgm3 on T. gondii vacuoles was also reported to occur in rare settings. Activation of the IRGs is thought to follow a GTP dependent cycle of IRG-IRG oligomerization. [15] The loading of the "pioneer" IRGs onto the vacuole is thought to greatly enhance the recruitment of additional IRGs in a cooperative fashion.

Pathogens have co-evolved unique mechanisms to interfere with different steps leading up to the association of the full complement of IRGs needed to constitute a vacuolar destructive complex. One such example was elucidated by infection with virulent and recombinant, avirulent strains of T. gondii. The intricate mechanism demonstrates a co-evolving interaction between the two species. Type I T. gondii rhoptry effector molecule Rop18, a serine-threonine kinase, was recently shown to selectively phosphorylate and inactivate the "pioneer" IRGs, thereby preventing their assembly, activation and destruction of the T. gondii vacuole within monocytes. [16] [17]

In addition to the role of IRGs in T. gondii infection, clearance of Mycobacterium, Mtb, was also shown to be adversely affected in mice lacking Irgm1, a key negative regulatory IRG. [10] The mechanism of clearance is thought to involve lipid interactions which aid in targeting the IRGs to Mtb containing phagosomes within macrophages. [18]

Figure 3: Following invasion by a number of obligate intracellular parasites, increases in interferon gamma lead to the increased synthesis of IRGs. The specific context and localization of the activated IRGs can lead to activation of several integrated cellular pathways resulting in either colonization of the host cell, death of the parasite, and / or death of the infected cell via one of the cell death pathways. A number of pathogens may also have effector molecules which can affect the activation of the cellular processes downstream of IRG activation. IRG activation following pathogen entry.jpg
Figure 3: Following invasion by a number of obligate intracellular parasites, increases in interferon gamma lead to the increased synthesis of IRGs. The specific context and localization of the activated IRGs can lead to activation of several integrated cellular pathways resulting in either colonization of the host cell, death of the parasite, and / or death of the infected cell via one of the cell death pathways. A number of pathogens may also have effector molecules which can affect the activation of the cellular processes downstream of IRG activation.

Another example of the role of IRGs in the mouse model of infection is demonstrated by the differential recruitment of IRGs which alter the outcome of Chlamydia trachomatis , a human adapted, versus Chlamydia muridarum , mouse adapted, inclusions following entry. C. trachomatis inclusions recruit a full repertoire of IRGs which aid in the inclusion elimination via fusion with lysosomes. [19] Regulation of IRG expression and activity in this model was found to be dependent both on levels of phospholipase C, cPLA2, as well as upstream IFN signaling. cPLA2 null mouse cells, when challenged with C. trachomatis, were less capable of clearing the pathogen compared to cells with proper cPLA2 expression. [20] The model highlights the co-evolution demonstrated by effector molecules of C. muridarum functioning to restrict accumulation of mouse IRGs on inclusions by modifying Irgb10, whereas the human pathogen is incapable of modifying the mouse IRG response. [19] This mechanism requires additional involvement of cellular autophagy machinery, which is in contrast to the necrotic pathway activation in T. gondii clearance. [17] A refined mechanism will require additional studies to elucidate the cooperation between the autophagy machinery and IRGs involved in the fusion of bacterial inclusions with the lysosome, as well as the specific bacterial effector molecules used to manipulate the coordinated actions of IRGs. [21]

In addition to the role of Irgm1 in parasite clearance, a cytoprotective role has been suggested in mature CD4+ T cells following exposure to IFNy in a TH1 response. [22] Irgm1 null mice were reported to develop pancytopenia following infection with both Mycobacterium avium and Trypanosoma cruzi . This phenotype was reversed when presented in a double IFNγ/Irgm1 knockout model. These studies have provided evidence that the role of IRGs may prove to be not only spatially and temporally regulated in a highly coordinated manner, but that they have context specific auxiliary roles outside of the traditional phagolysosome development and maturation. [22] [23]

IRGs and Humans

There are only three putative IRG genes in humans out of which IRGM is known to be an orthologue of mouse Irgm1. [4] There are four isoforms of IRGM (a-d). Unlike mouse IRGs, human IRGM isoforms are always expressed under the human retrovirus element ERV9 and are independent of the levels of IFNγ. IRGMb and d have a putative G5 (SAK) motif in their C terminal tail region while the other two isoforms do not. [24] IRGMd appears diffuse in the cytoplasm and be translocated to the mitochondria appearing as punctate dots. Additionally, it has been shown to bind to mitochondrial membrane lipid, cardiolipin and influence a change in the morphology of organelle. In general human IRGs have also been shown to affect several processes such as autophagy, mitochondrial fission, change in mitochondrial membrane potential and cell death.

Figure 4: Putative mechanism of action of Human IRGM. Under moderate to low expression levels, IRGMs constantly survey for signals such as pathogen entry, starvation or increase in levels of IFNg. Upon encounter with a pathogen, mitochondrial fission pathways are turned on leading to autophagy and microbial clearance promoting host cell survival. When highly expressed, IRGMd binds to the mitochondrial membrane bound lipid called cardiolipin and translocates into the mitochondria. This leads to depolarization, loss of mitochondrial membrane potential and stimulates pro-apoptotic Bax/Bak dependent host cell death. Putative mechaism of action of Human IRGM.jpg
Figure 4: Putative mechanism of action of Human IRGM. Under moderate to low expression levels, IRGMs constantly survey for signals such as pathogen entry, starvation or increase in levels of IFNγ. Upon encounter with a pathogen, mitochondrial fission pathways are turned on leading to autophagy and microbial clearance promoting host cell survival. When highly expressed, IRGMd binds to the mitochondrial membrane bound lipid called cardiolipin and translocates into the mitochondria. This leads to depolarization, loss of mitochondrial membrane potential and stimulates pro-apoptotic Bax/Bak dependent host cell death.

Human IRGM like its mouse counterpart, has been shown to play a role in autophagy, the mechanism of which is not fully understood. LC3 is a microtubule associated soluble protein found in mammalian tissues. Cytoplasmic proteins and organelles are engulfed by autophagosomes which convert LC3-I to LC3-II. The presence of LC3-II serves as a marker for autophagy and can be detected by immunofluorescence or immunoblotting. [25] IRGM helps to convert LC3-I to LC3-II in macrophages. [4] IRGMs have a dual role. When expressed at very low levels, they serve to protect against intracellular pathogens but when isoforms a, c and d are over expressed it leads to cell death and inflammation.

Studies indicate that lack of IRGMs are risk factors for Crohn's disease and tuberculosis. Humans use IRGM as a defense mechanism against the intracellular bacteria Mycobacterium tuberculosis . It has been found to be important in phagosome maturation and in reducing the number of intracellular mycobacteria with the help of other mitochondrial fission proteins such as DRP1 and FIs1. [4] Under certain conditions, mitochondrial fission and its associated proteins promote autophagy whereas mitochondrial fusion inhibits the same. Under autophagy inducing conditions, IRGM also increases ROS (Reactive Oxygen Species) production.

High levels of IRGMd triggers mitochondrial fission, leads to a loss of mitochondrial membrane potential and causes host cell death. Fission is also linked to mitochondrial Bax/Bak dependent apoptosis and IRGMd requires these proteins to be functional. Cell death by IRGM is independent of autophagy but depends on the above-mentioned pro-apoptotic factors instead. As a consequence of IRGM induced cell death, dying and necrotic cells release nuclear HMGB1, a pro-inflammatory alarmin, implicated in Crohn's disease. [24]

Related Research Articles

<span class="mw-page-title-main">Toxoplasmosis</span> Protozoan parasitic disease

Toxoplasmosis is a parasitic disease caused by Toxoplasma gondii, an apicomplexan. Infections with toxoplasmosis are associated with a variety of neuropsychiatric and behavioral conditions. Occasionally, people may have a few weeks or months of mild, flu-like illness such as muscle aches and tender lymph nodes. In a small number of people, eye problems may develop. In those with a weak immune system, severe symptoms such as seizures and poor coordination may occur. If a person becomes infected during pregnancy, a condition known as congenital toxoplasmosis may affect the child.

<i>Toxoplasma gondii</i> Type of protozoan parasite

Toxoplasma gondii is a parasitic protozoan that causes toxoplasmosis. Found worldwide, T. gondii is capable of infecting virtually all warm-blooded animals, but felids are the only known definitive hosts in which the parasite may undergo sexual reproduction.

<span class="mw-page-title-main">Autophagy</span> Cellular catabolic process in which cells digest parts of their own cytoplasm

Autophagy is the natural, conserved degradation of the cell that removes unnecessary or dysfunctional components through a lysosome-dependent regulated mechanism. It allows the orderly degradation and recycling of cellular components. Although initially characterized as a primordial degradation pathway induced to protect against starvation, it has become increasingly clear that autophagy also plays a major role in the homeostasis of non-starved cells. Defects in autophagy have been linked to various human diseases, including neurodegeneration and cancer, and interest in modulating autophagy as a potential treatment for these diseases has grown rapidly.

<span class="mw-page-title-main">Phagosome</span>

In cell biology, a phagosome is a vesicle formed around a particle engulfed by a phagocyte via phagocytosis. Professional phagocytes include macrophages, neutrophils, and dendritic cells (DCs).

<span class="mw-page-title-main">Interferon gamma</span> InterPro Family

Interferon gamma (IFN-γ) is a dimerized soluble cytokine that is the only member of the type II class of interferons. The existence of this interferon, which early in its history was known as immune interferon, was described by E. F. Wheelock as a product of human leukocytes stimulated with phytohemagglutinin, and by others as a product of antigen-stimulated lymphocytes. It was also shown to be produced in human lymphocytes. or tuberculin-sensitized mouse peritoneal lymphocytes challenged with Mantoux test (PPD); the resulting supernatants were shown to inhibit growth of vesicular stomatitis virus. Those reports also contained the basic observation underlying the now widely employed IFN-γ release assay used to test for tuberculosis. In humans, the IFN-γ protein is encoded by the IFNG gene.

<span class="mw-page-title-main">CXCL10</span> Mammalian protein found in Homo sapiens

C-X-C motif chemokine ligand 10 (CXCL10) also known as Interferon gamma-induced protein 10 (IP-10) or small-inducible cytokine B10 is an 8.7 kDa protein that in humans is encoded by the CXCL10 gene. C-X-C motif chemokine 10 is a small cytokine belonging to the CXC chemokine family.

<span class="mw-page-title-main">STAT1</span> Transcription factor and coding gene in humans

Signal transducer and activator of transcription 1 (STAT1) is a transcription factor which in humans is encoded by the STAT1 gene. It is a member of the STAT protein family.

Understanding of the antitumor immunity role of CD4+ T cells has grown substantially since the late 1990s. CD4+ T cells (mature T-helper cells) play an important role in modulating immune responses to pathogens and tumor cells, and are important in orchestrating overall immune responses.

<span class="mw-page-title-main">STAT2</span> Protein-coding gene in Homo sapiens

Signal transducer and activator of transcription 2 is a protein that in humans is encoded by the STAT2 gene. It is a member of the STAT protein family. This protein is critical to the biological response of type I interferons (IFNs). STAT2 sequence identity between mouse and human is only 68%.

<span class="mw-page-title-main">Vojo Deretic</span> American geneticist

Vojo Deretic, is distinguished professor and chair of the Department of Molecular Genetics and Microbiology at the University of New Mexico School of Medicine. Deretic was the founding director of the Autophagy, Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence. The AIM center promotes autophagy research nationally and internationally.

<span class="mw-page-title-main">ISG15</span> Protein-coding gene in the species Homo sapiens

Interferon-stimulated gene 15 (ISG15) is a 17 kDA secreted protein that in humans is encoded by the ISG15 gene. ISG15 is induced by type I interferon (IFN) and serves many functions, acting both as an extracellular cytokine and an intracellular protein modifier. The precise functions are diverse and vary among species but include potentiation of Interferon gamma (IFN-II) production in lymphocytes, ubiquitin-like conjugation to newly-synthesized proteins and negative regulation of the IFN-I response.

<span class="mw-page-title-main">GBP2</span> Protein-coding gene in the species Homo sapiens

Interferon-induced guanylate-binding protein 2 is a protein that in humans is encoded by the GBP2 gene. GBP2 is a gene related to the superfamily of large GTPases which can be induced mainly by interferon gamma.

Mitophagy is the selective degradation of mitochondria by autophagy. It often occurs to defective mitochondria following damage or stress. The process of mitophagy was first described over a hundred years ago by Margaret Reed Lewis and Warren Harmon Lewis. Ashford and Porter used electron microscopy to observe mitochondrial fragments in liver lysosomes by 1962, and a 1977 report suggested that "mitochondria develop functional alterations which would activate autophagy." The term "mitophagy" was in use by 1998.

<span class="mw-page-title-main">Toll-like receptor 11</span>

Toll-like receptor 11 (TLR11) is a protein that in mice and rats is encoded by the gene TLR11, whereas in humans it is represented by a pseudogene. TLR11 belongs to the toll-like receptor (TLR) family and the interleukin-1 receptor/toll-like receptor superfamily. In mice, TLR11 has been shown to recognise (bacterial) flagellin and (eukaryotic) profilin present on certain microbes, it helps propagate a host immune response. TLR11 plays a fundamental role in both the innate and adaptive immune responses, through the activation of Tumor necrosis factor-alpha, the Interleukin 12 (IL-12) response, and Interferon-gamma (IFN-gamma) secretion. TLR11 mounts an immune response to multiple microbes, including Toxoplasma gondii, Salmonella species, and uropathogenic E. coli, and likely many other species due to the highly conserved nature of flagellin and profilin.

<span class="mw-page-title-main">IRGM</span> Protein-coding gene in the species Homo sapiens

Immunity-related GTPase family M protein (IRGM), also known as interferon-inducible protein 1 (IFI1), is an enzyme that in humans is encoded by the IRGM gene.

<span class="mw-page-title-main">Interferon-inducible GTPase 5</span> Protein-coding gene in the species Homo sapiens

Interferon-inducible GTPase 5 also known as immunity-related GTPase cinema 1 (IRGC1) is an enzyme that in humans is coded by the IRGC gene. It is predicted to behave like other proteins in the p47-GTPase-like and IRG families. It is most expressed in the testis.

<span class="mw-page-title-main">Stimulator of interferon genes</span> Protein-coding gene in the species Homo sapiens

Stimulator of interferon genes (STING), also known as transmembrane protein 173 (TMEM173) and MPYS/MITA/ERIS is a protein that in humans is encoded by the STING1 gene.

The cGAS–STING pathway is a component of the innate immune system that functions to detect the presence of cytosolic DNA and, in response, trigger expression of inflammatory genes that can lead to senescence or to the activation of defense mechanisms. DNA is normally found in the nucleus of the cell. Localization of DNA to the cytosol is associated with tumorigenesis, viral infection, and invasion by some intracellular bacteria. The cGAS – STING pathway acts to detect cytosolic DNA and induce an immune response.

<span class="mw-page-title-main">Rubicon (protein)</span> Human protein involved in autophagy regulation

Rubicon is a protein that in humans is encoded by the RUBCN gene. Rubicon is one of the few known negative regulators of autophagy, a cellular process that degrades unnecessary or damaged cellular components. Rubicon is recruited to its sites of action through interaction with the small GTPase Rab7, and impairs the autophagosome-lysosome fusion step of autophagy through inhibition of PI3KC3-C2.

References

  1. 1 2 Li G, Zhang J, Sun Y, Wang H, Wang Y (2009). "The evolutionarily dynamic IFN-inducible GTPase proteins play conserved immune functions in vertebrates and cephalochordates". Mol Biol Evol. 26 (7): 1619–30. doi: 10.1093/molbev/msp074 . PMID   19369598.
  2. Huang S, Yuan S, Guo L, Yu Y, Li J, Wu T, et al. (2008). "Genomic analysis of the immune gene repertoire of amphioxus reveals extraordinary innate complexity and diversity". Genome Res. 18 (7): 1112–26. doi:10.1101/gr.069674.107. PMC   2493400 . PMID   18562681.
  3. 1 2 3 Bekpen C, Hunn JP, Rohde C, Parvanova I, Guethlein L, Dunn DM, et al. (2005). "The interferon-inducible p47 (IRG) GTPases in vertebrates: loss of the cell autonomous resistance mechanism in the human lineage". Genome Biol. 6 (11): R92. doi: 10.1186/gb-2005-6-11-r92 . PMC   1297648 . PMID   16277747.
  4. 1 2 3 4 Singh SB, Davis AS, Taylor GA, Deretic V (2006). "Human IRGM induces autophagy to eliminate intracellular mycobacteria". Science. 313 (5792): 1438–41. Bibcode:2006Sci...313.1438S. doi:10.1126/science.1129577. PMID   16888103. S2CID   2274272.
  5. Gilly M, Wall R (1992). "The IRG-47 gene is IFN-gamma induced in B cells and encodes a protein with GTP-binding motifs". J Immunol. 148 (10): 3275–81. doi: 10.4049/jimmunol.148.10.3275 . PMID   1578148. S2CID   10508833.
  6. Taylor GA, Jeffers M, Largaespada DA, Jenkins NA, Copeland NG, Woude GF (1996). "Identification of a novel GTPase, the inducibly expressed GTPase, that accumulates in response to IFNγ". J Biol Chem. 271 (34): 20399–405. doi: 10.1074/jbc.271.34.20399 . PMID   8702776.
  7. Taylor GA, Collazo CM, Yap GS, Nguyen K, Gregorio TA, Taylor LS, et al. (2000). "Pathogen-specific loss of host resistance in mice lacking the IFN-gamma-inducible gene IGTP". Proc Natl Acad Sci U S A. 97 (2): 751–5. Bibcode:2000PNAS...97..751T. doi: 10.1073/pnas.97.2.751 . PMC   15402 . PMID   10639151.
  8. Feng CG, Collazo-Custodio CM, Eckhaus M, Hieny S, Belkaid Y, Elkins K, et al. (2004). "Mice deficient in LRG-47 display increased susceptibility to mycobacterial infection associated with the induction of lymphopenia". J Immunol. 172 (2): 1163–8. doi: 10.4049/jimmunol.172.2.1163 . PMID   14707092.
  9. Zeng J, Parvanova IA, Howard JC (2009). "A dedicated promoter drives constitutive expression of the cell-autonomous immune resistance GTPase, Irga6 (IIGP1) in mouse liver". PLOS ONE. 4 (8): e6787. Bibcode:2009PLoSO...4.6787Z. doi: 10.1371/journal.pone.0006787 . PMC   2848866 . PMID   20368812.
  10. 1 2 MacMicking JD, Taylor GA, McKinney JD (2003). "Immune control of tuberculosis by IFN-gamma-inducible LRG-47". Science. 302 (5645): 654–9. Bibcode:2003Sci...302..654M. doi:10.1126/science.1088063. PMID   14576437. S2CID   83944695.
  11. Pawlowski N, Khaminets A, Hunn JP, Papic N, Schmidt A, Uthaiah RC, et al. (2011). "The activation mechanism of Irga6, an interferon-inducible GTPase contributing to mouse resistance against Toxoplasma gondii". BMC Biol. 9: 7. doi: 10.1186/1741-7007-9-7 . PMC   3042988 . PMID   21276251.
  12. 1 2 Khaminets A, Hunn JP, Könen-Waisman S, Zhao YO, Preukschat D, Coers J, et al. (2010). "Coordinated loading of IRG resistance GTPases on to the Toxoplasma gondii parasitophorous vacuole". Cell Microbiol. 12 (7): 939–61. doi:10.1111/j.1462-5822.2010.01443.x. PMC   2901525 . PMID   20109161.
  13. Zhao YO, Könen-Waisman S, Taylor GA, Martens S, Howard JC (2010). "Localisation and mislocalisation of the interferon-inducible immunity-related GTPase, Irgm1 (LRG-47) in mouse cells". PLOS ONE. 5 (1): e8648. Bibcode:2010PLoSO...5.8648Z. doi: 10.1371/journal.pone.0008648 . PMC   2799677 . PMID   20072621.
  14. Martens S, Sabel K, Lange R, Uthaiah R, Wolf E, Howard JC (2004). "Mechanisms regulating the positioning of mouse p47 resistance GTPases LRG-47 and IIGP1 on cellular membranes: retargeting to plasma membrane induced by phagocytosis". J Immunol. 173 (4): 2594–606. doi: 10.4049/jimmunol.173.4.2594 . PMID   15294976.
  15. Uthaiah RC, Praefcke GJ, Howard JC, Herrmann C (2003). "IIGP1, an interferon-gamma-inducible 47-kDa GTPase of the mouse, showing cooperative enzymatic activity and GTP-dependent multimerization". J Biol Chem. 278 (31): 29336–43. doi: 10.1074/jbc.M211973200 . PMID   12732635.
  16. Fentress SJ, Behnke MS, Dunay IR, Mashayekhi M, Rommereim LM, Fox BA, et al. (2010). "Phosphorylation of immunity-related GTPases by a Toxoplasma gondii-secreted kinase promotes macrophage survival and virulence". Cell Host Microbe. 8 (6): 484–95. doi:10.1016/j.chom.2010.11.005. PMC   3013631 . PMID   21147463.
  17. 1 2 Zhao YO, Khaminets A, Hunn JP, Howard JC (2009). "Disruption of the Toxoplasma gondii parasitophorous vacuole by IFNgamma-inducible immunity-related GTPases (IRG proteins) triggers necrotic cell death". PLOS Pathog. 5 (2): e1000288. doi: 10.1371/journal.ppat.1000288 . PMC   2629126 . PMID   19197351.
  18. Tiwari S, Choi HP, Matsuzawa T, Pypaert M, MacMicking JD (2009). "Targeting of the GTPase Irgm1 to the phagosomal membrane via PtdIns(3,4)P(2) and PtdIns(3,4,5)P(3) promotes immunity to mycobacteria". Nat Immunol. 10 (8): 907–17. doi:10.1038/ni.1759. PMC   2715447 . PMID   19620982.
  19. 1 2 Coers J, Bernstein-Hanley I, Grotsky D, Parvanova I, Howard JC, Taylor GA, et al. (2008). "Chlamydia muridarum evades growth restriction by the IFN-gamma-inducible host resistance factor Irgb10". J Immunol. 180 (9): 6237–45. doi: 10.4049/jimmunol.180.9.6237 . PMID   18424746.
  20. Vignola MJ, Kashatus DF, Taylor GA, Counter CM, Valdivia RH (2010). "cPLA2 regulates the expression of type I interferons and intracellular immunity to Chlamydia trachomatis". J Biol Chem. 285 (28): 21625–35. doi: 10.1074/jbc.M110.103010 . PMC   2898388 . PMID   20452986.
  21. Al-Zeer MA, Al-Younes HM, Braun PR, Zerrahn J, Meyer TF (2009). "IFN-gamma-inducible Irga6 mediates host resistance against Chlamydia trachomatis via autophagy". PLOS ONE. 4 (2): e4588. Bibcode:2009PLoSO...4.4588A. doi: 10.1371/journal.pone.0004588 . PMC   2643846 . PMID   19242543.
  22. 1 2 Feng CG, Zheng L, Jankovic D, Báfica A, Cannons JL, Watford WT, et al. (2008). "The immunity-related GTPase Irgm1 promotes the expansion of activated CD4+ T cell populations by preventing interferon-gamma-induced cell death". Nat Immunol. 9 (11): 1279–87. doi:10.1038/ni.1653. PMC   2580721 . PMID   18806793.
  23. Feng CG, Zheng L, Lenardo MJ, Sher A (2009). "Interferon-inducible immunity-related GTPase Irgm1 regulates IFN gamma-dependent host defense, lymphocyte survival and autophagy". Autophagy. 5 (2): 232–4. doi:10.4161/auto.5.2.7445. PMC   2749220 . PMID   19066452.
  24. 1 2 Singh SB, Ornatowski W, Vergne I, Naylor J, Delgado M, Roberts E, et al. (2010). "Human IRGM regulates autophagy and cell-autonomous immunity functions through mitochondria". Nat Cell Biol. 12 (12): 1154–65. doi:10.1038/ncb2119. PMC   2996476 . PMID   21102437.
  25. Tanida I, Ueno T, Kominami E (2008). "LC3 and Autophagy". Autophagosome and Phagosome. Methods Mol Biol. Vol. 445. pp. 77–88. doi:10.1007/978-1-59745-157-4_4. ISBN   978-1-58829-853-9. PMID   18425443.