Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Immunity ; 53(2): 398-416.e8, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814028

ABSTRACT

Paneth cells are the primary source of C-type lysozyme, a ß-1,4-N-acetylmuramoylhydrolase that enzymatically processes bacterial cell walls. Paneth cells are normally present in human cecum and ascending colon, but are rarely found in descending colon and rectum; Paneth cell metaplasia in this region and aberrant lysozyme production are hallmarks of inflammatory bowel disease (IBD) pathology. Here, we examined the impact of aberrant lysozyme production in colonic inflammation. Targeted disruption of Paneth cell lysozyme (Lyz1) protected mice from experimental colitis. Lyz1-deficiency diminished intestinal immune responses to bacterial molecular patterns and resulted in the expansion of lysozyme-sensitive mucolytic bacteria, including Ruminococcus gnavus, a Crohn's disease-associated pathobiont. Ectopic lysozyme production in colonic epithelium suppressed lysozyme-sensitive bacteria and exacerbated colitis. Transfer of R. gnavus into Lyz1-/- hosts elicited a type 2 immune response, causing epithelial reprograming and enhanced anti-colitogenic capacity. In contrast, in lysozyme-intact hosts, processed R. gnavus drove pro-inflammatory responses. Thus, Paneth cell lysozyme balances intestinal anti- and pro-inflammatory responses, with implications for IBD.


Subject(s)
Clostridiales/immunology , Colitis, Ulcerative/pathology , Muramidase/genetics , Muramidase/metabolism , Paneth Cells/metabolism , Animals , Clostridiales/genetics , Colitis, Ulcerative/microbiology , Crohn Disease/pathology , Female , Gastrointestinal Microbiome/genetics , Goblet Cells/cytology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , STAT6 Transcription Factor/genetics
2.
Nat Immunol ; 17(4): 356-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27002843

ABSTRACT

Innate cells are responsible for the rapid recognition of infection and mediate essential mechanisms of pathogen elimination, and also facilitate adaptive immune responses. We review here the numerous intricate interactions among innate cells that initiate protective immunity. The efficient eradication of pathogens depends on the coordinated actions of multiple cells, including innate cells and epithelial cells. Rather than acting as isolated effector cells, innate cells are in constant communication with other responding cells of the immune system, locally and distally. These interactions are critically important for the efficient control of primary infections as well for the development of 'trained' innate cells that facilitate the rapid elimination of homologous or heterologous infections.


Subject(s)
Adaptive Immunity/immunology , Cytokines/immunology , Immunity, Innate/immunology , Infections/immunology , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Animals , Basophils/immunology , Eosinophils/immunology , Humans , Macrophages/immunology , Mast Cells/immunology , Monocytes/immunology , Neutrophils/immunology
3.
Nat Immunol ; 14(11): 1118-1126, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24145791

ABSTRACT

Helminth infections are ubiquitous worldwide and can trigger potent immune responses that differ from and potentially antagonize host protective responses to microbial pathogens. In this Review we focus on the three main killers in infectious disease-AIDS, tuberculosis and malaria-and critically assesses whether helminths adversely influence host control of these diseases. We also discuss emerging concepts for how M2 macrophages and helminth-modulated dendritic cells can potentially influence the protective immune response to concurrent infections. Finally, we present evidence advocating for more efforts to determine how and to what extent helminths interfere with the successful control of specific concurrent coinfections.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Dendritic Cells/immunology , Helminthiasis/immunology , Macrophages/immunology , Malaria/immunology , Tuberculosis/immunology , Acquired Immunodeficiency Syndrome/epidemiology , Acquired Immunodeficiency Syndrome/virology , Africa/epidemiology , Animals , Asia/epidemiology , Coinfection , Dendritic Cells/microbiology , Dendritic Cells/parasitology , Dendritic Cells/virology , Helminthiasis/epidemiology , Helminthiasis/parasitology , Helminths/immunology , Host-Parasite Interactions , Humans , Latin America/epidemiology , Macrophages/microbiology , Macrophages/parasitology , Macrophages/virology , Malaria/epidemiology , Malaria/parasitology , Tuberculosis/epidemiology , Tuberculosis/microbiology
4.
J Immunol ; 210(1): 5-11, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36542831

ABSTRACT

Growth differentiation factor 15 (GDF-15) is a cytokine that is widely used as a biomarker for the severity of diverse disease states. It also has been shown to play a protective role after tissue injury and to promote a negative energy balance during obesity and diabetes. In addition to its metabolic effects, GDF-15 also regulates the host's immune responses to infectious and noninfectious diseases. GDF-15 can suppress a type 1 and, in contrast, promote a type 2 inflammatory response. In this brief review, we discuss how GDF-15 affects the effector function and recruitment of immune cells, the pathways that induce its expression, and the diverse mechanisms by which it is regulated during inflammation and infection. We further highlight outstanding questions that should be the focus of future investigations in this emerging field.


Subject(s)
Diabetes Mellitus , Growth Differentiation Factor 15 , Humans , Growth Differentiation Factor 15/metabolism , Biomarkers/metabolism , Inflammation , Immunity
5.
Brain Behav Immun ; 116: 24-33, 2024 02.
Article in English | MEDLINE | ID: mdl-38013040

ABSTRACT

Infections are often accompanied by weight loss caused by alterations in host behavior and metabolism, also known as sickness behaviors. Recent studies have revealed that sickness behaviors can either promote or impede survival during infections depending on factors such as the type of infectious pathogen. Nevertheless, we have an incomplete understanding of the underlying mechanisms of sickness behaviors. Furthermore, although the host immune responses to infections have long been known to contribute to the induction of sickness behaviors, recent studies have identified emerging cytokines that are also key regulators of host metabolism during infection and inflammation, such as growth differentiation factor 15 (GDF-15). GDF-15 is a distant member of the TGF-ß superfamily that causes weight loss by suppressing appetite and food consumption and causing emesis. These effects require activation of neurons that express the only known GDF-15 receptor, the GFRAL receptor. GDF-15 also functions in the periphery including the induction of ketogenesis and immunoregulation. Nevertheless, the functions and regulation of GDF-15 during live infections is not yet known. Murine infection with avirulent Toxoplasma gondii is an established model to understand infection-induced weight loss. Past studies have determined that acute T. gondii infection causes weight loss due to diminished food consumption and increased energy expenditure through unknown mechanisms. Additionally, our lab previously demonstrated that T. gondii causes upregulation in serum GDF-15 in an IFN-γ-dependent manner during the post-acute phase of the infection. In this study, we interrogated the in-vivo functions and immune regulation of GDF-15 during Toxoplasma gondii infection. First, we found that in wild-type mice, acute T. gondii infection caused a significant weight loss that is preceded by elevation of serum levels of IFN-γ and GDF-15. To determine whether IFN-γ regulates GDF-15, we neutralized IFN-γ on days 5 and 6 and measured GDF-15 on day 7 and found that serum but not tissue levels of GDF-15 decreased after IFN-γ neutralization. Additionally, exogenous IFN-γ was sufficient to elevate serum GDF-15 in the absence of infection. Next, we compared the outcomes of T. gondii infection between WT and Gdf15-/- mice. We observed that the weight trajectories were declining in WT mice while they were increasing in Gdf15-/-mice during the acute phase of the infection. This difference in trajectories extended throughout the chronic infection resulting to an overall weight loss relative to initial weights in WT mice but not Gdf15-/-mice. Then, we determined that GDF-15 is not essential for survival and immunoregulation during T. gondii infection. We also demonstrated that GDF-15 is required for the induction of FGF21, stress-induced cytokine with prominent roles in regulating host metabolism. Finally, we discovered a cytokine cascade IFN-γ-GDF-15-FGF21 that is likely involved in the regulation of host metabolism. Overall, our study provides evidence that IFN-γ contributes to the regulation of host metabolism during infection by inducing GDF-15 and FGF21. GDF-15 orchestrates changes in host metabolism that supports the host immune response in clearing the infection. These physiological alterations induce FGF21, which in turn, orchestrates the adaptive responses to the effects of GDF-15, which can be detrimental when protracted.


Subject(s)
Fibroblast Growth Factors , Toxoplasma , Toxoplasmosis , Animals , Mice , Growth Differentiation Factor 15/genetics , Interferon-gamma/metabolism , Cytokines
6.
J Immunol ; 209(11): 2160-2171, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36426972

ABSTRACT

More than 2 billion people worldwide are infected with helminths. Thus, it is possible for individuals to experience concomitant infection with helminth and intracellular microbes. Although the helminth-induced type 2 response can suppress type 1 proinflammatory responses required for the immunity against intracellular pathogens in the context of a coinfection, conflicting evidence suggest that helminth infection can enhance antimicrobial immunity. Using a coinfection model with the intestinal helminth Heligmosomoides polygyrus followed by infection with Toxoplasma gondii in Mus Musculus, we showed that the complex and dynamic effect of helminth infection is highly suppressive during the innate phase (days 0-3) of T. gondii infection and less stringent during the acute phase (d10). Helminth coinfection had a strong suppressive effect on the neutrophil, monocytic, and early IFN-γ/IL-12 responses. The IFN-γ response was later restored by compensatory production from T cells despite decreased effector differentiation of T. gondii-specific CD8 T cells. In accordance with the attenuated IFN-γ response, parasite loads were elevated during the acute phase (d10) of T. gondii infection but were transiently controlled by the compensatory T cell response. Unexpectedly, 40% of helminth-coinfected mice exhibited a sustained weight loss phenotype during the postacute phase (d14-18) that was not associated with T. gondii outgrowth, indicating that coinfection led to decreased disease tolerance during T. gondii infection. Our work uncovers the dynamic nature of the helminth immunomodulatory effects on concomitant infections or immune responses and unveils a loss of disease tolerance phenotype triggered by coinfection with intestinal helminth.


Subject(s)
Coinfection , Nematospiroides dubius , Toxoplasma , Toxoplasmosis , Animals , Mice , Immune Tolerance
7.
J Immunol ; 207(6): 1507-1512, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34400524

ABSTRACT

Resistance and tolerance are vital for survivability of the host-pathogen relationship. Virulence during Toxoplasma infection in mice is mediated by parasite kinase-dependent antagonism of IFN-γ-induced host resistance. Whether avirulence requires expression of parasite factors that induce host tolerance mechanisms or is a default status reflecting the absence of resistance-interfering factors is not known. In this study, we present evidence that avirulence in Toxoplasma requires parasite engagement of the scavenger receptor CD36. CD36 promotes macrophage tropism but is dispensable for the development of resistance mechanisms. Instead CD36 is critical for re-establishing tissue homeostasis and survival following the acute phase of infection. The CD36-binding capacity of T. gondii strains is negatively controlled by the virulence factor, ROP18. Thus, the absence of resistance-interfering virulence factors and the presence of tolerance-inducing avirulence factors are both required for long-term host-pathogen survival.


Subject(s)
CD36 Antigens/deficiency , CD36 Antigens/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/parasitology , Toxoplasma/metabolism , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/immunology , Animals , CD36 Antigens/genetics , CHO Cells , Cricetulus , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Protozoan Proteins/metabolism , RAW 264.7 Cells , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Animal/parasitology , Virulence/genetics , Virulence Factors/metabolism
8.
Infect Immun ; 90(8): e0020522, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35913173

ABSTRACT

The role of specific host cell surface receptors during Toxoplasma gondii invasion of host cells is poorly defined. Here, we interrogated the role of the well-known malarial invasion receptor, basigin, in T. gondii infection of astrocytes. We found that primary astrocytes express two members of the BASIGIN (BSG) immunoglobulin family, basigin and embigin, but did not express neuroplastin. Antibody blockade of either basigin or embigin caused a significant reduction of parasite infectivity in astrocytes. The specific role of basigin during T. gondii invasion was further examined using a mouse astrocytic cell line (C8-D30), which exclusively expresses basigin. CRISPR-mediated deletion of basigin in C8-D30 cells resulted in decreased T. gondii infectivity. T. gondii replication and invasion efficiency were not altered by basigin deficiency, but parasite attachment to astrocytes was markedly reduced. We also conducted a proteomic screen to identify T. gondii proteins that interact with basigin. Toxoplasma-encoded cyclophilins, the protein 14-3-3, and protein disulfide isomerase (TgPDI) were among the putative basigin-ligands identified. Recombinant TgPDI produced in E. coli bound to basigin and pretreatment of tachyzoites with a PDI inhibitor decreased parasite attachment to host cells. Finally, mutagenesis of the active site cysteines of TgPDI abolished enzyme binding to basigin. Thus, basigin and its related immunoglobulin family members may represent host receptors that mediate attachment of T. gondii to diverse cell types.


Subject(s)
Toxoplasma , Toxoplasmosis , Basigin , Escherichia coli , Humans , Proteomics
9.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Article in English | MEDLINE | ID: mdl-32421753

ABSTRACT

Anti-helminth responses require robust type 2 cytokine production that simultaneously promotes worm expulsion and initiates the resolution of helminth-induced wounds and hemorrhaging. However, how infection-induced changes in hematopoiesis contribute to these seemingly distinct processes remains unknown. Recent studies have suggested the existence of a hematopoietic progenitor with dual mast cell-erythrocyte potential. Nonetheless, whether and how these progenitors contribute to host protection during an active infection remains to be defined. Here, we employed single cell RNA-sequencing and identified that the metabolic enzyme, carbonic anhydrase (Car) 1 marks a predefined bone marrow-resident hematopoietic progenitor cell (HPC) population. Next, we generated a Car1-reporter mouse model and found that Car1-GFP positive progenitors represent bipotent mast cell/erythrocyte precursors. Finally, we show that Car1-expressing HPCs simultaneously support mast cell and erythrocyte responses during Trichinella spiralis infection. Collectively, these data suggest that mast cell/erythrocyte precursors are mobilized to promote type 2 cytokine responses and alleviate helminth-induced blood loss, developmentally linking these processes. Collectively, these studies reveal unappreciated hematopoietic events initiated by the host to combat helminth parasites and provide insight into the evolutionary pressure that may have shaped the developmental relationship between mast cells and erythrocytes.


Subject(s)
Erythroid Precursor Cells/immunology , Erythropoiesis/immunology , Mast Cells/immunology , Mastocytosis/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Carbonic Anhydrase I/genetics , Carbonic Anhydrase I/immunology , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/pathology , Female , Mast Cells/parasitology , Mast Cells/pathology , Mastocytosis/genetics , Mastocytosis/pathology , Mice , Mice, Transgenic , Trichinellosis/genetics , Trichinellosis/pathology
10.
Immunity ; 36(6): 904-6, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22749350

ABSTRACT

After acute inflammation or infection, tissue-resident macrophages are replaced by inflammatory DCs and effector macrophages that arise from circulating monocytes. In this issue of Immunity, Goldszmid et al. (2012) demonstrate a central role for NK cell-derived IFN-γ in signaling and facilitating this transition.

11.
J Immunol ; 200(1): 186-195, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180487

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are the major producers of IFN-α, an antiviral cytokine involved in immunomodulation and control of HIV type 1 replication, whereas Toxoplasma gondii is a life-threatening opportunistic infection in AIDS patients. During infection with HIV type 1, human pDCs decrease in circulation and remaining pDC produce lower amounts of IFN-α in response to viral stimulation. In this study, we investigated the impact of coinfection with T. gondii on the innate virus-directed responses of human pDCs. Using intracellular flow cytometry and fluorescence microscopy, we determined that T. gondii invaded but did not induce IFN-α or TNF-α in human pDC. However, T. gondii inhibited IFN-α and TNF-α produced in response to HSV and HIV, thus functionally inactivating pDC. IFN-α production was inhibited only in cells infected by T. gondii, which inhibited neither uptake of GFP-HSV nor localization of TLR9 in CD71+ endosomes, directing us to investigate downstream events. Using imaging flow cytometry, we found that both T. gondii and IL-10 inhibited virus-induced nuclear translocation, but not phosphorylation, of IFN response factor 7. Blockade of IFN response factor 7 nuclear translocation and inhibition of the IFN-α response was partially reversed by a deficiency in the T. gondii-derived ROP16 kinase, known to directly phosphorylate STAT3, a critical mediator of IL-10's anti-inflammatory effects. Taken together, our results indicate that T. gondii suppresses pDC activation by mimicking IL-10's regulatory effects through an ROP16 kinase-dependent mechanism. Our findings further imply a convergent mechanism of inhibition of TLR signaling by T. gondii and IL-10 and suggest potential negative consequences of HIV/T. gondii coinfection.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/immunology , Interleukin-10/metabolism , Opportunistic Infections/immunology , Protein-Tyrosine Kinases/metabolism , Protozoan Proteins/metabolism , Toxoplasma/immunology , Toxoplasmosis/immunology , Cell Differentiation , Cells, Cultured , Coinfection , Dendritic Cells/parasitology , Humans , Immunity, Innate , Immunomodulation , Interferon Regulatory Factor-7/metabolism , Interferon-alpha/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
J Immunol ; 198(2): 634-639, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27956529

ABSTRACT

Concurrent helminth infection potently inhibits T cell immunity; however, whether helminthes prevent T cell priming or skew clonal recruitment and effector differentiation is not known. Using coinfection with two natural mouse pathogens, Heligmosomoides polygyrus and Toxoplasma gondii, to investigate the negative impact of helminthes on the CD8 T cell response, we demonstrate helminth-induced suppression of IL-12-dependent differentiation of killer-like receptor G1+ effector CD8 T cells and IFN-γ production. Nevertheless, reversal of helminth suppression of the innate IL-12 response of CD8α+ dendritic cells, which occurred in STAT6-deficient mice, was not sufficient to normalize CD8 T cell differentiation. Instead, a combined deficiency in IL-4 and IL-10 was required to reverse the negative effects of helminth coinfection on the CD8 T cell response. Monoclonal T. gondii-specific CD8 T cells adoptively transferred into coinfected mice recapitulated the spectrum of helminth-induced effects on the polyclonal CD8 T response, indicating the lack of requirement for clonal skewing.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Coinfection/immunology , Helminthiasis/immunology , Lymphocyte Activation/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Disease Models, Animal , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , Toxoplasma/immunology
13.
J Immunol ; 194(1): 210-22, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25416805

ABSTRACT

Beta-catenin signaling has recently been tied to the emergence of tolerogenic dendritic cells (DCs). In this article, we demonstrate a novel role for beta-catenin in directing DC subset development through IFN regulatory factor 8 (IRF8) activation. We found that splenic DC precursors express beta-catenin, and DCs from mice with CD11c-specific constitutive beta-catenin activation upregulated IRF8 through targeting of the Irf8 promoter, leading to in vivo expansion of IRF8-dependent CD8a+, plasmacytoid, and CD103+ CD11b2 DCs. beta-catenin­stabilized CD8a+ DCs secreted elevated IL-12 upon in vitro microbial stimulation, and pharmacological beta-catenin inhibition blocked this response in wild-type cells. Upon infections with Toxoplasma gondii and vaccinia virus, mice with stabilized DC beta-catenin displayed abnormally high Th1 and CD8+ T lymphocyte responses, respectively. Collectively, these results reveal a novel and unexpected function for beta-catenin in programming DC differentiation toward subsets that orchestrate proinflammatory immunity to infection.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Inflammation/immunology , Interferon Regulatory Factors/genetics , beta Catenin/immunology , Animals , Antigens, CD/immunology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CD11c Antigen/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Enzyme Activation , Female , Integrin alpha Chains/immunology , Interferon Regulatory Factors/immunology , Interleukin-12/biosynthesis , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasite Load , Promoter Regions, Genetic , Pyrimidinones/pharmacology , Receptors, Cell Surface/genetics , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Th1 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Vaccinia/immunology , Vaccinia virus/immunology , beta Catenin/antagonists & inhibitors , beta Catenin/biosynthesis
14.
Proc Natl Acad Sci U S A ; 111(17): 6437-42, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733931

ABSTRACT

Unlike most intracellular pathogens that gain access into host cells through endocytic pathways, Toxoplasma gondii initiates infection at the cell surface by active penetration through a moving junction and subsequent formation of a parasitophorous vacuole. Here, we describe a noncanonical pathway for T. gondii infection of macrophages, in which parasites are initially internalized through phagocytosis, and then actively invade from within a phagosomal compartment to form a parasitophorous vacuole. This phagosome to vacuole invasion (PTVI) pathway may represent an intermediary link between the endocytic and the penetrative routes for host cell entry by intracellular pathogens. The PTVI pathway is preferentially used by avirulent strains of T. gondii and confers an infectious advantage over virulent strains for macrophage tropism.


Subject(s)
Macrophages/parasitology , Phagosomes/parasitology , Toxoplasma/pathogenicity , Animals , Cell Line , Macrophages/pathology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Phagocytosis , Phagosomes/pathology , Phagosomes/ultrastructure , Toxoplasma/ultrastructure , Toxoplasmosis/parasitology , Toxoplasmosis/pathology , Tropism , Vacuoles/parasitology , Vacuoles/pathology , Vacuoles/ultrastructure
16.
Trends Parasitol ; 40(6): 449-451, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762372

ABSTRACT

Polymeric guanylate-binding proteins (GBPs) physically dismember the vacuole membrane formed by Toxoplasma gondii while nitric oxide (NO) poisons and inhibits parasite replication within interferon (IFN)-γ activated macrophages. Zhao et al. report a novel mechanism for synergy between these classical microbicidal and microbistatic effectors in cell-autonomous immunity to the intracellular parasites.


Subject(s)
Toxoplasma , Toxoplasma/immunology , Nitric Oxide/metabolism , Animals , Humans , GTP-Binding Proteins/immunology , GTP-Binding Proteins/metabolism , Macrophages/immunology , Macrophages/parasitology
17.
J Infect Dis ; 205(1): 152-61, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21990421

ABSTRACT

Recent studies have underscored physiological and pathophysiological roles for the tryptophan-degrading enzyme indolamine 2,3-dioxygenase (IDO) in immune counterregulation. However, IDO was first recognized as an antimicrobial effector, restricting tryptophan availability to Toxoplasma gondii and other pathogens in vitro. The biological relevance of these findings came under question when infectious phenotypes were not forthcoming in IDO-deficient mice. The recent discovery of an IDO homolog, IDO-2, suggested that the issue deserved reexamination. IDO inhibition during murine toxoplasmosis led to 100% mortality, with increased parasite burdens and no evident effects on the immune response. Similar studies revealed a counterregulatory role for IDO during leishmaniasis (restraining effector immune responses and parasite clearance), and no evident role for IDO in herpes simplex virus type 1 (HSV-1) infection. Thus, IDO plays biologically important roles in the host response to diverse intracellular infections, but the dominant nature of this role--antimicrobial or immunoregulatory--is pathogen-specific.


Subject(s)
Herpes Simplex/enzymology , Herpesvirus 1, Human , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Leishmaniasis, Cutaneous/immunology , Toxoplasmosis, Animal/immunology , Animals , Female , Herpes Simplex/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Leishmaniasis, Cutaneous/enzymology , Male , Mice , Mice, Inbred C57BL , Toxoplasmosis, Animal/enzymology , Tryptophan/analogs & derivatives , Tryptophan/metabolism
18.
Trends Parasitol ; 39(1): 10-11, 2023 01.
Article in English | MEDLINE | ID: mdl-36470783

ABSTRACT

Toxoplasma gondii exploits the migratory properties of monocytes and dendritic cells to promote tissue dissemination. Recently, ten Hoeve et al. reported that the parasite effector protein GRA28 conspires with host chromatin modifiers to confer dendritic cell-like features that convert sessile macrophages into migratory cells that transport infection to distal organs.


Subject(s)
Toxoplasma , Macrophages/parasitology , Dendritic Cells/parasitology
19.
Cell Host Microbe ; 31(12): 1952-1953, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38096787

ABSTRACT

Globally, cryptosporidiosis is a leading cause of childhood diarrheal disease and is a major risk factor for malnutrition and impairment of growth and cognitive development. In this issue of Cell Host & Microbe, Maradana et al. identify a target for dietary enhancement of innate immune defenses against cryptosporidiosis.


Subject(s)
Cryptosporidiosis , Cryptosporidium , Malnutrition , Humans , Diarrhea , Diet
20.
J Exp Med ; 203(9): 2063-71, 2006 Sep 04.
Article in English | MEDLINE | ID: mdl-16940170

ABSTRACT

Apicomplexan protozoan pathogens avoid destruction and establish a replicative niche within host cells by forming a nonfusogenic parasitophorous vacuole (PV). Here we present evidence for lysosome-mediated degradation of Toxoplasma gondii after invasion of macrophages activated in vivo. Pathogen elimination was dependent on the interferon gamma inducible-p47 GTPase, IGTP, required PI3K activity, and was preceded by PV membrane indentation, vesiculation, disruption, and, surprisingly, stripping of the parasite plasma membrane. Denuded parasites were enveloped in autophagosome-like vacuoles, which ultimately fused with lysosomes. These observations outline a series of mechanisms used by effector cells to redirect the fate of a classically nonfusogenic intracellular pathogen toward a path of immune elimination.


Subject(s)
Autophagy/physiology , Cell Membrane/metabolism , Macrophages , Toxoplasma/metabolism , Vacuoles/metabolism , Animals , Cell Membrane/parasitology , Cell Membrane/ultrastructure , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Immunohistochemistry , Interferon-gamma/immunology , Lysosomes/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Macrophages/parasitology , Membrane Fusion/physiology , Mice , Mice, Inbred Strains , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Toxoplasma/immunology , Vacuoles/parasitology , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL