Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 12: 755961, 2021.
Article in English | MEDLINE | ID: mdl-34867993

ABSTRACT

Non-canonical inflammasome activation by mouse caspase-11 (or human CASPASE-4/5) is crucial for the clearance of certain gram-negative bacterial infections, but can lead to severe inflammatory damage. Factors that promote non-canonical inflammasome activation are well recognized, but less is known about the mechanisms underlying its negative regulation. Herein, we identify that the caspase-11 inflammasome in mouse and human macrophages (Mϕ) is negatively controlled by the zinc (Zn2+) regulating protein, metallothionein 3 (MT3). Upon challenge with intracellular lipopolysaccharide (iLPS), Mϕ increased MT3 expression that curtailed the activation of caspase-11 and its downstream targets caspase-1 and interleukin (IL)-1ß. Mechanistically, MT3 increased intramacrophage Zn2+ to downmodulate the TRIF-IRF3-STAT1 axis that is prerequisite for caspase-11 effector function. In vivo, MT3 suppressed activation of the caspase-11 inflammasome, while caspase-11 and MT3 synergized in impairing antibacterial immunity. The present study identifies an important yin-yang relationship between the non-canonical inflammasome and MT3 in controlling inflammation and immunity to gram-negative bacteria.


Subject(s)
Caspases/immunology , Gram-Negative Bacterial Infections/immunology , Inflammasomes/immunology , Macrophages/immunology , Metallothionein 3/immunology , Zinc/immunology , Animals , Caspases/metabolism , Gram-Negative Bacterial Infections/metabolism , Humans , Inflammasomes/metabolism , Macrophages/metabolism , Metallothionein 3/metabolism , Mice , Mice, Inbred C57BL , Zinc/metabolism
2.
Cell Rep ; 27(13): 3873-3886.e7, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31242420

ABSTRACT

Alternatively activated (M2) macrophages promote wound healing but weaken antimicrobial defenses. The mechanisms that enforce macrophage divergence and dictate the phenotypic and metabolic characteristics of M2 macrophages remain elusive. We show that alternative activation with interleukin (IL)-4 induces expression of metallothionein 3 (MT3) that regulates macrophage polarization and function. MT3 was requisite for metabolic reprograming in IL-4-stimulated macrophages or M(IL-4) macrophages to promote mitochondrial respiration and suppress glycolysis. MT3 fostered an M(IL-4) phenotype, suppressed hypoxia inducible factor (HIF)1α activation, and thwarted the emergence of a proinflammatory M1 program in macrophages. MT3 deficiency augmented macrophage plasticity, resulting in enhanced interferon γ (IFNγ) responsiveness and a dampened M(IL-4) phenotype. Thus, MT3 programs the phenotype and metabolic fate of M(IL-4) macrophages.


Subject(s)
Glycolysis , Macrophage Activation , Macrophages/metabolism , Nerve Tissue Proteins/metabolism , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Metallothionein 3 , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
3.
Int J Mol Sci ; 18(10)2017 Oct 23.
Article in English | MEDLINE | ID: mdl-29065550

ABSTRACT

Metallothioneins (MTs) are a family of metal-binding proteins virtually expressed in all organisms including prokaryotes, lower eukaryotes, invertebrates and mammals. These proteins regulate homeostasis of zinc (Zn) and copper (Cu), mitigate heavy metal poisoning, and alleviate superoxide stress. In recent years, MTs have emerged as an important, yet largely underappreciated, component of the immune system. Innate and adaptive immune cells regulate MTs in response to stress stimuli, cytokine signals and microbial challenge. Modulation of MTs in these cells in turn regulates metal ion release, transport and distribution, cellular redox status, enzyme function and cell signaling. While it is well established that the host strictly regulates availability of metal ions during microbial pathogenesis, we are only recently beginning to unravel the interplay between metal-regulatory pathways and immunological defenses. In this perspective, investigation of mechanisms that leverage the potential of MTs to orchestrate inflammatory responses and antimicrobial defenses has gained momentum. The purpose of this review, therefore, is to illumine the role of MTs in immune regulation. We discuss the mechanisms of MT induction and signaling in immune cells and explore the therapeutic potential of the MT-Zn axis in bolstering immune defenses against pathogens.


Subject(s)
Immune System/metabolism , Infections/metabolism , Metallothionein/immunology , Animals , Cytokines , Humans , Metallothionein/metabolism , Metals/metabolism , Signal Transduction
4.
Cell Rep ; 16(12): 3232-3246, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653687

ABSTRACT

Alternative activation of macrophages promotes wound healing but weakens antimicrobial defenses against intracellular pathogens. The mechanisms that suppress macrophage function to create a favorable environment for pathogen growth remain elusive. We show that interleukin (IL)-4 triggers a metallothionein 3 (MT3)- and Zn exporter SLC30A4-dependent increase in the labile Zn(2+) stores in macrophages and that intracellular pathogens can exploit this increase in Zn to survive. IL-4 regulates this pathway by shuttling extracellular Zn into macrophages and by activating cathepsins that act on MT3 to release bound Zn. We show that IL-4 can modulate Zn homeostasis in both human monocytes and mice. In vivo, MT3 can repress macrophage function in an M2-polarizing environment to promote pathogen persistence. Thus, MT3 and SLC30A4 dictate the size of the labile Zn(2+) pool and promote the survival of a prototypical intracellular pathogen in M2 macrophages.


Subject(s)
Cation Transport Proteins/metabolism , Host-Pathogen Interactions/physiology , Interleukin-4/metabolism , Macrophages/microbiology , Nerve Tissue Proteins/metabolism , Zinc/metabolism , Animals , Cation Transport Proteins/immunology , Histoplasmosis/immunology , Histoplasmosis/metabolism , Humans , Interleukin-4/immunology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Metallothionein 3 , Mice , Nerve Tissue Proteins/immunology
5.
J Immunol ; 197(5): 1864-76, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27465530

ABSTRACT

Zinc (Zn) is an essential metal for development and maintenance of both the innate and adaptive compartments of the immune system. Zn homeostasis impacts maturation of dendritic cells (DCs) that are important in shaping T cell responses. The mechanisms by which Zn regulates the tolerogenic phenotype of DCs remain largely unknown. In this study, we investigated the effect of Zn on DC phenotype and the generation of Foxp3(+) regulatory T cells (Tregs) using a model of Histoplasma capsulatum fungal infection. Exposure of bone marrow-derived DCs to Zn in vitro induced a tolerogenic phenotype by diminishing surface MHC class II (MHCII) and promoting the tolerogenic markers, programmed death-ligand (PD-L)1, PD-L2, and the tryptophan degrading enzyme, IDO. Zn triggered tryptophan degradation by IDO and kynurenine production by DCs and strongly suppressed the proinflammatory response to stimulation by TLR ligands. In vivo, Zn supplementation and subsequent H. capsulatum infection supressed MHCII on DCs, enhanced PD-L1 and PD-L2 expression on MHCII(lo) DCs, and skewed the Treg-Th17 balance in favor of Foxp3(+) Tregs while decreasing Th17 cells. Thus, Zn shapes the tolerogenic potential of DCs in vitro and in vivo and promotes Tregs during fungal infection.


Subject(s)
Dendritic Cells/drug effects , Histoplasmosis/immunology , Immune Tolerance , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Zinc/pharmacology , Animals , Bone Marrow Cells/drug effects , Dendritic Cells/immunology , Genes, MHC Class II/immunology , Histoplasma/immunology , Histoplasma/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Lymphocyte Activation , Mice , Phenotype , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Tryptophan/metabolism , Zinc/physiology
6.
Cell Host Microbe ; 19(3): 361-74, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26922990

ABSTRACT

Systemic fungal infections trigger marked immune-regulatory disturbances, but the mechanisms are poorly understood. We report that the pathogenic yeast of Blastomyces dermatitidis elaborates dipeptidyl-peptidase IVA (DppIVA), a close mimic of the mammalian ectopeptidase CD26, which modulates critical aspects of hematopoiesis. We show that, like the mammalian enzyme, fungal DppIVA cleaved C-C chemokines and GM-CSF. Yeast producing DppIVA crippled the recruitment and differentiation of monocytes and prevented phagocyte activation and ROS production. Silencing fungal DppIVA gene expression curtailed virulence and restored recruitment of CCR2(+) monocytes, generation of TipDC, and phagocyte killing of yeast. Pharmacological blockade of DppIVA restored leukocyte effector functions and stemmed infection, while addition of recombinant DppIVA to gene-silenced yeast enabled them to evade leukocyte defense. Thus, fungal DppIVA mediates immune-regulatory disturbances that underlie invasive fungal disease. These findings reveal a form of molecular piracy by a broadly conserved aminopeptidase during disease pathogenesis.


Subject(s)
Aminopeptidases/metabolism , Blastomyces/enzymology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Immune Evasion , Immune Tolerance , Immunity, Innate/drug effects , Virulence Factors/metabolism , Animals , Biological Mimicry , Blastomyces/pathogenicity , Chemokines/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Gene Silencing , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Mice , Microbial Viability , Monocytes/immunology , Phagocytosis , Reactive Oxygen Species/metabolism , Sequence Homology, Amino Acid , Virulence Factors/genetics
7.
Arch Biochem Biophys ; 611: 66-78, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-26921502

ABSTRACT

The importance of Zn ions (Zn) in regulating development and functions of the immune system is well established. However, recent years have witnessed a surge in our knowledge of how immune cells choreograph Zn regulatory mechanisms to combat the persistence of pathogenic microbes. Myeloid and lymphoid populations manipulate intracellular and extracellular Zn metabolism via Zn binding proteins and transporters in response to immunological signals and infection. Rapid as well as delayed changes in readily exchangeable Zn, also known as free Zn and the Zn proteome are crucial in determining activation of immune cells, cytokine responses, signaling and nutritional immunity. Recent studies have unearthed distinctive Zn modulatory mechanisms employed by specialized immune cells and necessitate an understanding of the Zn handling behavior in immune responses to infection. The focus of this review, therefore, stems from novel revelations of Zn intoxication, sequestration and signaling roles deployed by different immune cells, with an emphasis on innate immunity, to challenge microbial parasitization and cope with pathogen insult.


Subject(s)
Host-Pathogen Interactions , Immune System , Immunity, Innate , Zinc/immunology , Animals , Anti-Infective Agents/chemistry , Cytokines/metabolism , Homeostasis , Humans , Inflammation , Leukocyte L1 Antigen Complex/chemistry , Macrophages/metabolism , Manganese/chemistry , Neutrophils/metabolism , Oxidative Stress , Signal Transduction
8.
Immunity ; 39(4): 697-710, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24138881

ABSTRACT

Macrophages possess numerous mechanisms to combat microbial invasion, including sequestration of essential nutrients, like zinc (Zn). The pleiotropic cytokine granulocyte macrophage-colony stimulating factor (GM-CSF) enhances antimicrobial defenses against intracellular pathogens such as Histoplasma capsulatum, but its mode of action remains elusive. We have found that GM-CSF-activated infected macrophages sequestered labile Zn by inducing binding to metallothioneins (MTs) in a STAT3 and STAT5 transcription-factor-dependent manner. GM-CSF upregulated expression of Zn exporters, Slc30a4 and Slc30a7; the metal was shuttled away from phagosomes and into the Golgi apparatus. This distinctive Zn sequestration strategy elevated phagosomal H⁺ channel function and triggered reactive oxygen species generation by NADPH oxidase. Consequently, H. capsulatum was selectively deprived of Zn, thereby halting replication and fostering fungal clearance. GM-CSF mediated Zn sequestration via MTs in vitro and in vivo in mice and in human macrophages. These findings illuminate a GM-CSF-induced Zn-sequestration network that drives phagocyte antimicrobial effector function.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Histoplasma/immunology , Histoplasmosis/metabolism , Macrophages, Peritoneal/immunology , Superoxides/metabolism , Zinc/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Gene Expression Regulation , Golgi Apparatus/drug effects , Golgi Apparatus/immunology , Golgi Apparatus/microbiology , Histoplasma/drug effects , Histoplasmosis/immunology , Histoplasmosis/microbiology , Host-Pathogen Interactions , Humans , Macrophage Activation , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/microbiology , Metallothionein/genetics , Metallothionein/immunology , Mice , Mice, Transgenic , NADPH Oxidases/genetics , NADPH Oxidases/immunology , Phagosomes/drug effects , Phagosomes/immunology , Phagosomes/microbiology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , Superoxides/immunology , Zinc/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...