Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37683721

ABSTRACT

BACKGROUND: Factor XII (FXII) is a multifunctional protease capable of activating thrombotic and inflammatory pathways. FXII has been linked to thrombosis in extracorporeal membrane oxygenation (ECMO), but the role of FXII in ECMO-induced inflammatory complications has not been studied. We used novel gene-targeted FXII- deficient rats to evaluate the role of FXII in ECMO-induced thromboinflammation. METHODS: FXII-deficient (FXII-/-) Sprague-Dawley rats were generated using CRISPR/Cas9. A minimally invasive venoarterial (VA) ECMO model was used to compare wild-type (WT) and FXII-/- rats in 2 separate experimental cohorts: rats placed on ECMO without pharmacologic anticoagulation and rats anticoagulated with argatroban. Rats were maintained on ECMO for 1 hour or until circuit failure occurred. Comparisons were made with unchallenged rats and rats that underwent a sham surgical procedure without ECMO. RESULTS: FXII-/- rats were maintained on ECMO without pharmacologic anticoagulation with low resistance throughout the 1-hour experiment. In contrast, WT rats placed on ECMO without anticoagulation developed thrombotic circuit failure within 10 minutes. Argatroban provided a means to maintain WT and FXII-/- rats on ECMO for the 1-hour time frame without thrombotic complications. Analyses of these rats demonstrated that ECMO resulted in increased neutrophil migration into the liver that was significantly blunted by FXII deficiency. ECMO also resulted in increases in high molecular weight kininogen cleavage and complement activation that were abrogated by genetic deletion of FXII. CONCLUSIONS: FXII initiates hemostatic system activation and key inflammatory sequelae in ECMO, suggesting that therapies targeting FXII could limit both thromboembolism and inopportune inflammatory complications in this setting.

2.
J Immunol ; 208(4): 793-798, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35101895

ABSTRACT

Metabolomics analyses suggest changes in amino acid abundance, particularly l-arginine (L-ARG), occur in patients with tuberculosis. Immune cells require L-ARG to fuel effector functions following infection. We have previously described an L-ARG synthesis pathway in immune cells; however, its role in APCs has yet to be uncovered. Using a coculture system with mycobacterial-specific CD4+ T cells, we show APC L-ARG synthesis supported T cell viability and proliferation, and activated T cells contained APC-derived L-ARG. We hypothesize that APCs supply L-ARG to support T cell activation under nutrient-limiting conditions. This work expands the current model of APC-T cell interactions and provides insight into the effects of nutrient availability in immune cells.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Arginine/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/immunology , Animals , Arginine/biosynthesis , Argininosuccinic Aciduria/etiology , Argininosuccinic Aciduria/metabolism , Biological Transport , Biomarkers , Cell Proliferation , Cell Survival/immunology , Flow Cytometry , Immunophenotyping , Lymphocyte Activation/genetics , Mice , Mice, Transgenic
3.
mBio ; 12(6): e0271021, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34749531

ABSTRACT

Hypoxia-inducible factor 1α (HIF-1α) regulates the immunometabolic phenotype of macrophages, including the orchestration of inflammatory and antimicrobial processes. Macrophages deficient in HIF-1α produce excessive quantities of the anti-inflammatory cytokine interleukin 10 (IL-10) during infection with the intracellular fungal pathogen Histoplasma capsulatum (R. A. Fecher, M. C. Horwath, D. Friedrich, J. Rupp, G. S. Deepe, J Immunol 197:565-579, 2016, https://doi.org/10.4049/jimmunol.1600342). Thus, the macrophage fails to become activated in response to proinflammatory cytokines and remains the intracellular niche of the pathogen. Here, we identify the tricarboxylic acid (TCA) cycle metabolite fumarate as the driver of IL-10 during macrophage infection with H. capsulatum in the absence of HIF-1α. Accumulation of fumarate reduced expression of a HIF-1α-dependent microRNA (miRNA), miR-27a, known to mediate decay of Il10 mRNA. Inhibition of fumarate accrual in vivo limited IL-10 and fungal growth. Our data demonstrate the critical role of HIF-1α in shaping appropriate TCA cycle activity in response to infection and highlight the consequences of a dysregulated immunometabolic response. IMPORTANCE Histoplasma capsulatum and related Histoplasma species are intracellular fungal pathogens endemic to broad regions of the globe, including the Americas, Africa, and Asia. While most infections resolve with mild or no symptoms, failure of the host to control fungal growth produces severe disease. Previously, we reported that loss of a key transcriptional regulator, hypoxia-inducible factor 1α (HIF-1α), in macrophages led to a lethal failure to control growth of Histoplasma (R. A. Fecher, M. C. Horwath, D. Friedrich, J. Rupp, G. S. Deepe, J Immunol 197:565-579, 2016, https://doi.org/10.4049/jimmunol.1600342). Inhibition of phagocyte activation due to excessive interleukin 10 by HIF-1α-deficient macrophages drove this outcome. In this study, we demonstrate that HIF-1α maintains contextually appropriate TCA cycle metabolism within Histoplasma-infected macrophages. The absence of HIF-1α results in excessive fumarate production that alters miRNA-27a regulation of interleukin-10. HIF-1α thus preserves the capacity of macrophages to transition from a permissive intracellular niche to the site of pathogen killing.


Subject(s)
Fumarates/metabolism , Histoplasma/physiology , Histoplasmosis/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-10/metabolism , Macrophages/microbiology , MicroRNAs/metabolism , Animals , Citric Acid Cycle , Histoplasma/genetics , Histoplasmosis/genetics , Histoplasmosis/microbiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-10/genetics , Macrophages/metabolism , Mice , MicroRNAs/genetics
4.
Cell Host Microbe ; 29(12): 1744-1756.e5, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34678170

ABSTRACT

Interactions between the microbiota and mammalian host are essential for defense against infection, but the microbial-derived cues that mediate this relationship remain unclear. Here, we find that intestinal epithelial cell (IEC)-associated commensal bacteria, segmented filamentous bacteria (SFB), promote early protection against the pathogen Citrobacter rodentium, independent of CD4+ T cells. SFB induced histone modifications in IECs at sites enriched for retinoic acid receptor motifs, suggesting that SFB may enhance defense through retinoic acid (RA). Consistent with this, inhibiting RA signaling suppressed SFB-induced protection. Intestinal RA levels were elevated in SFB mice, despite the inhibition of mammalian RA production, indicating that SFB directly modulate RA. Interestingly, RA was produced by intestinal bacteria, and the loss of bacterial-intrinsic aldehyde dehydrogenase activity decreased the RA levels and increased infection. These data reveal RA as an unexpected microbiota-derived metabolite that primes innate defense and suggests that pre- and probiotic approaches to elevate RA could prevent or combat infections.


Subject(s)
Bacteria/metabolism , Intestinal Diseases/metabolism , Symbiosis , Tretinoin/metabolism , Animals , Bacillus cereus , Bifidobacterium bifidum , CD4-Positive T-Lymphocytes , Citrobacter rodentium , Epithelial Cells , Histone Code , Host Microbial Interactions , Intestinal Diseases/microbiology , Male , Mice , Mice, Inbred C57BL , Microbiota , Nitric Oxide , Signal Transduction
5.
Front Immunol ; 12: 653571, 2021.
Article in English | MEDLINE | ID: mdl-34054815

ABSTRACT

Macrophages are indispensable immune cells tasked at eliminating intracellular pathogens. Mycobacterium tuberculosis (Mtb), one of the most virulent intracellular bacterial pathogens known to man, infects and resides within macrophages. While macrophages can be provoked by extracellular stimuli to inhibit and kill Mtb bacilli, these host defense mechanisms can be blocked by limiting nutritional metabolites, such as amino acids. The amino acid L-arginine has been well described to enhance immune function, especially in the context of driving macrophage nitric oxide (NO) production in mice. In this study, we aimed to establish the necessity of L-arginine on anti-Mtb macrophage function independent of NO. Utilizing an in vitro system, we identified that macrophages relied on NO for only half of their L-arginine-mediated host defenses and this L-arginine-mediated defense in the absence of NO was associated with enhanced macrophage numbers and viability. Additionally, we observed macrophage glycolysis to be driven by both L-arginine and mechanistic target of rapamycin (mTOR), and inhibition of glycolysis or mTOR reduced macrophage control of Mtb as well as macrophage number and viability in the presence of L-arginine. Our data underscore L-arginine as an essential nutrient for macrophage function, not only by fueling anti-mycobacterial NO production, but also as a central regulator of macrophage metabolism and additional host defense mechanisms.


Subject(s)
Arginine/metabolism , Dietary Supplements , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/diet therapy , Animals , Arginine/administration & dosage , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/metabolism , Argininosuccinate Synthase/genetics , Argininosuccinate Synthase/metabolism , Cell Survival , Disease Models, Animal , Humans , Macrophage Activation , Macrophages/metabolism , Mice , Mice, Knockout , Nitric Oxide/metabolism , Primary Cell Culture , RAW 264.7 Cells , Tuberculosis/immunology , Tuberculosis/microbiology
6.
Front Immunol ; 11: 628432, 2020.
Article in English | MEDLINE | ID: mdl-33633745

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a leading cause of death worldwide. Despite decades of research, there is still much to be uncovered regarding the immune response to Mtb infection. Here, we summarize the current knowledge on anti-Mtb immunity, with a spotlight on immune cell amino acid metabolism. Specifically, we discuss L-arginine and L-tryptophan, focusing on their requirements, regulatory roles, and potential use as adjunctive therapy in TB patients. By continuing to uncover the immune cell contribution during Mtb infection and how amino acid utilization regulates their functions, it is anticipated that novel host-directed therapies may be developed and/or refined, helping to eradicate TB.


Subject(s)
Arginine , Mycobacterium tuberculosis , Tryptophan , Tuberculosis , Arginine/immunology , Arginine/metabolism , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Tryptophan/immunology , Tryptophan/metabolism , Tuberculosis/immunology , Tuberculosis/metabolism
7.
Front Immunol ; 11: 607957, 2020.
Article in English | MEDLINE | ID: mdl-33391278

ABSTRACT

Tuberculosis has been associated with increased risk of atherosclerotic cardiovascular disease. To examine whether mycobacterial infection exacerbates atherosclerosis development in experimental conditions, we infected low-density lipoprotein receptor knockout (Ldlr-/-) mice with Mycobacterium bovis Bacille-Calmette-Guérin (BCG), an attenuated strain of the Mycobacterium tuberculosis complex. Twelve-week old male Ldlr-/- mice were infected with BCG (0.3-3.0x106 colony-forming units) via the intranasal route. Mice were subsequently fed a western-type diet containing 21% fat and 0.2% cholesterol for up to 16 weeks. Age-matched uninfected Ldlr-/- mice fed with an identical diet served as controls. Atherosclerotic lesions in aorta were examined using Oil Red O staining. Changes induced by BCG infection on the immunophenotyping profile of circulating T lymphocytes and monocytes were assessed using flow cytometry. BCG infection increased atherosclerotic lesions in en face aorta after 8 weeks (plaque ratio; 0.021±0.01 vs. 0.013±0.01; p = 0.011) and 16 weeks (plaque ratio, 0.15±0.13 vs. 0.06±0.02; p = 0.003). No significant differences in plasma cholesterol or triglyceride levels were observed between infected and uninfected mice. Compared to uninfected mice, BCG infection increased systemic CD4/CD8 T cell ratio and the proportion of Ly6Clow non-classical monocytes at weeks 8 and 16. Aortic plaque ratios correlated with CD4/CD8 T cell ratios (Spearman's rho = 0.498; p = 0.001) and the proportion of Ly6Clow non-classical monocytes (Spearman's rho = 0.629; p < 0.001) at week 16. In conclusion, BCG infection expanded the proportion of CD4+ T cell and Ly6Clow monocytes, and aggravated atherosclerosis formation in the aortas of hyperlipidemic Ldlr-/- mice. Our results indicate that mycobacterial infection is capable of enhancing atherosclerosis development.


Subject(s)
Aorta/microbiology , Aortic Diseases/microbiology , Atherosclerosis/microbiology , Mycobacterium bovis/pathogenicity , Plaque, Atherosclerotic , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Diet, High-Fat , Disease Models, Animal , Disease Progression , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/microbiology , Receptors, LDL/genetics , Receptors, LDL/metabolism
8.
J Thromb Haemost ; 18(1): 91-103, 2020 01.
Article in English | MEDLINE | ID: mdl-31539206

ABSTRACT

BACKGROUND: Protease-activated receptor-1 (PAR-1) plays a major role in multiple disease processes, including colitis. Understanding the mechanisms coupling PAR-1 to disease pathogenesis is complicated by the fact that PAR-1 is broadly expressed across multiple cell types. OBJECTIVE: Determine the specific contributions of PAR-1 expressed by macrophages and colonic enterocytes to infectious colitis. METHODS: Mice carrying a conditional PAR-1 allele were generated and bred to mice expressing Cre recombinase in a myeloid- (PAR-1ΔM ) or enterocyte-specific (PAR-1ΔEPI ) fashion. Citrobacter rodentium colitis pathogenesis was analyzed in mice with global PAR-1 deletion (PAR-1-/- ) and cell type-specific deletions. RESULTS: Constitutive deletion of PAR-1 had no significant impact on weight loss, crypt hypertrophy, crypt abscess formation, or leukocyte infiltration in Citrobacter colitis. However, colonic shortening was significantly blunted in infected PAR-1-/- mice, and these animals exhibited decreased local levels of IL-1ß, IL-22, IL-6, and IL-17A. In contrast, infected PAR-1ΔM mice lost less weight and had fewer crypt abscesses relative to controls. PAR-1ΔM mice had diminished CD3+ T cell infiltration into colonic tissue, but macrophage and CD4+ T cell infiltration were similar to controls. Also contrasting results in global knockouts, PAR-1ΔM mice exhibited lower levels of IL-1ß, but not Th17-related cytokines (ie, IL-22, IL-6, IL-17A). Infected PAR-1ΔEPI mice exhibited increased crypt hypertrophy and crypt abscess formation, but local cytokine elaboration was similar to controls. CONCLUSIONS: These studies reveal complex, cell type-specific roles for PAR-1 in modulating the immune response to Citrobacter colitis that are not readily apparent in analyses limited to mice with global PAR-1 deficiency.


Subject(s)
Colitis , Receptor, PAR-1 , Animals , Citrobacter rodentium , Colitis/genetics , Colitis/microbiology , Enterobacteriaceae Infections , Mice , Mice, Inbred C57BL , Receptor, PAR-1/genetics , Th17 Cells
9.
J Immunol ; 202(6): 1747-1754, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30710047

ABSTRACT

Immunonutrition as a therapeutic approach is rapidly gaining interest in the fight against infection. Targeting l-arginine metabolism is intriguing, considering this amino acid is the substrate for antimicrobial NO production by macrophages. The importance of l-arginine during infection is supported by the finding that inhibiting its synthesis from its precursor l-citrulline blunts host defense. During the first few weeks following pulmonary mycobacterial infection, we found a drastic increase in l-citrulline in the lung, even though serum concentrations were unaltered. This correlated with increased gene expression of the l-citrulline-generating (i.e., iNOS) and l-citrulline-using (i.e., Ass1) enzymes in key myeloid populations. Eliminating l-arginine synthesis from l-citrulline in myeloid cells via conditional deletion of either Ass1 or Asl resulted in increased Mycobacterium bovis bacillus Calmette-Guérin and Mycobacterium tuberculosis H37Rv burden in the lungs compared with controls. Our data illustrate the necessity of l-citrulline metabolism for myeloid defense against mycobacterial infection and highlight the potential for host-directed therapy against mycobacterial disease targeting this nutrient and/or its metabolic pathway.


Subject(s)
Arginine/metabolism , Citrulline/metabolism , Mycobacterium Infections/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Animals , Arginine/immunology , Citrulline/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mycobacterium Infections/metabolism , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism
10.
Front Immunol ; 10: 2893, 2019.
Article in English | MEDLINE | ID: mdl-31921154

ABSTRACT

Obesity is a prevalent predisposing factor to non-alcoholic fatty liver disease (NAFLD), the most common chronic liver disease in the developed world. NAFLD spectrum of disease involves progression from steatosis (NAFL), to steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma (HCC). Despite clinical and public health significance, current FDA approved therapies for NAFLD are lacking in part due to insufficient understanding of pathogenic mechanisms driving disease progression. The etiology of NAFLD is multifactorial. The induction of both systemic and tissue inflammation consequential of skewed immune cell metabolic state, polarization, tissue recruitment, and activation are central to NAFLD progression. Here, we review the current understanding of the above stated cellular and molecular processes that govern macrophage contribution to NAFLD pathogenesis and how adipose tissue and liver crosstalk modulates macrophage function. Notably, the manipulation of such events may lead to the development of new therapies for NAFLD.


Subject(s)
Disease Susceptibility , Macrophages/immunology , Macrophages/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Biomarkers , Cell Movement , Cell Plasticity/immunology , Cytokines/metabolism , Disease Management , Disease Models, Animal , Energy Metabolism , Humans , Inflammation Mediators/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/therapy , Signal Transduction
11.
Front Immunol ; 8: 1561, 2017.
Article in English | MEDLINE | ID: mdl-29201027

ABSTRACT

Activation, recruitment, and effector function of T lymphocytes are essential for control of mycobacterial infection. These processes are tightly regulated in T cells by the availability of l-arginine within the microenvironment. In turn, mycobacterial infection dampens T cell responsiveness through arginase induction in myeloid cells, promoting sequestration of l-arginine within the local milieu. Here, we show T cells can replenish intracellular l-arginine through metabolism of l-citrulline to mediate inflammatory function, allowing anti-mycobacterial T cells to overcome arginase-mediated suppression. Furthermore, T cell l-citrulline metabolism is necessary for accumulation of CD4+ T cells at the site of infection, suggesting this metabolic pathway is involved during anti-mycobacterial T cell immunity in vivo. Together, these findings establish a contribution for l-arginine synthesis by T cells during mycobacterial infection, and implicate l-citrulline as a potential immuno-nutrient to modulate host immunity.

12.
J Pathol ; 242(4): 463-475, 2017 08.
Article in English | MEDLINE | ID: mdl-28497484

ABSTRACT

The CD44 gene encodes several protein isoforms due to alternative splicing and post translational modifications. Given that CD44 variant isoform 9 (CD44v9) is expressed within Spasmolytic Polypeptide/TFF2-Expressing Metaplasia (SPEM) glands during repair, CD44v9 may be play a funcitonal role during the process of regeneration of the gastric epithelium. Here we hypothesize that CD44v9 marks a regenerative cell lineage responsive to infiltrating macrophages during regeneration of the gastric epithelium. Ulcers were induced in CD44-deficient (CD44KO) and C57BL/6 (BL6) mice by a localized application of acetic acid to the serosal surface of the stomach. Gastric organoids expressing CD44v9 were derived from mouse stomachs and transplanted at the ulcer site of CD44KO mice. Ulcers, CD44v9 expression, proliferation and histology were measured 1, 3, 5 and 7-days post-injury. Human-derived gastric organoids were generated from stomach tissue collected from elderly (>55 years) or young (14-20 years) patients. Organoids were transplanted into the stomachs of NOD scid gamma (NSG) mice at the site of injury. Gastric injury was induced in NRG-SGM3 (NRGS) mice harboring human-derived immune cells (hnNRGS) and the immune profile anlayzed by CyTOF. CD44v9 expression emerged within regenerating glands the ulcer margin in response to injury. While ulcers in BL6 mice healed within 7-days post-injury, CD44KO mice exhibited loss of repair and epithelial regeneration. Ulcer healing was promoted in CD44KO mice by transplanted CD55v9-expressing gastric organoids. NSG mice exhibited loss of CD44v9 expression and gastric repair. Transplantation of human-derived gastric organoids from young, but not aged stomachs promoted repair in NSG mouse stomachs in response to injury. Finally, compared to NRGS mice, huNRGS animals exhibited reduced ulcer sizes, an infiltration of human CD162+ macrophages and an emergence of CD44v9 expression in SPEM. Thus, during repair of the gastic epithelium CD44v9 emerges within a regenerative cell lineage that coincides with macrophage inflitration within the injured mucosa. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Gastric Mucosa/physiology , Hyaluronan Receptors/genetics , Regeneration/physiology , Stomach Ulcer/metabolism , Adolescent , Age Factors , Aged , Animals , Cells, Cultured , Gastric Mucosa/pathology , Genetic Variation/physiology , Humans , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/physiology , Macrophages/physiology , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Middle Aged , Organoids/cytology , Organoids/transplantation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Regeneration/genetics , Stomach Ulcer/genetics , Stomach Ulcer/pathology , Wound Healing/physiology , Young Adult
13.
J Biol Chem ; 292(1): 15-30, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27903651

ABSTRACT

Modulation of T cell proliferation and function by immunoregulatory myeloid cells are an essential means of preventing self-reactivity and restoring tissue homeostasis. Consumption of amino acids such as arginine and tryptophan by immunoregulatory macrophages is one pathway that suppresses local T cell proliferation. Using a reduced complexity in vitro macrophage-T cell co-culture system, we show that macrophage arginase-1 is the only factor required by M2 macrophages to block T cells in G1, and this effect is mediated by l-arginine elimination rather than metabolite generation. Tracking how T cells adjust their metabolism when deprived of arginine revealed the significance of macrophage-mediated arginine deprivation to T cells. We found mTORC1 activity was unaffected in the initial G1 block. After 2 days of arginine deprivation, mTORC1 activity declined paralleling a selective down-regulation of SREBP target gene expression, whereas mRNAs involved in glycolysis, gluconeogenesis, and T cell activation were unaffected. Cell cycle arrest was reversible at any point by exogenous arginine, suggesting starved T cells remain poised awaiting nutrients. Arginine deprivation-induced cell cycle arrest was mediated in part by Rictor/mTORC2, providing evidence that this nutrient recognition pathway is a central component of how T cells measure environmental arginine.


Subject(s)
Arginine/metabolism , Carrier Proteins/metabolism , Cell Cycle Checkpoints/immunology , Cell Proliferation , Multiprotein Complexes/metabolism , Myeloid Cells/immunology , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Coculture Techniques , Immune Tolerance , Immunosuppression Therapy , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 2 , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , T-Lymphocytes/metabolism
15.
Cancer Cell ; 30(3): 377-390, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27622331

ABSTRACT

Effective cancer immunotherapy requires overcoming immunosuppressive tumor microenvironments. We found that local nitric oxide (NO) production by tumor-infiltrating myeloid cells is important for adoptively transferred CD8(+) cytotoxic T cells to destroy tumors. These myeloid cells are phenotypically similar to inducible nitric oxide synthase (NOS2)- and tumor necrosis factor (TNF)-producing dendritic cells (DC), or Tip-DCs. Depletion of immunosuppressive, colony stimulating factor 1 receptor (CSF-1R)-dependent arginase 1(+) myeloid cells enhanced NO-dependent tumor killing. Tumor elimination via NOS2 required the CD40-CD40L pathway. We also uncovered a strong correlation between survival of colorectal cancer patients and NOS2, CD40, and TNF expression in their tumors. Our results identify a network of pro-tumor factors that can be targeted to boost cancer immunotherapies.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Nitric Oxide Synthase Type II/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Arginase/biosynthesis , Arginase/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/biosynthesis , Tumor Microenvironment , Tumor Necrosis Factor-alpha/biosynthesis
16.
Semin Immunopathol ; 38(2): 139-52, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26490974

ABSTRACT

Tuberculosis (TB) granulomas are compact, organized agglomerations of infected and uninfected macrophages, T cells, neutrophils, and other immune cells. Within the granuloma, several unique metabolic adaptations occur to modify the behavior of immune cells, potentially favoring bacterial persistence balanced with protection against immunopathology. These include the induction of arginase-1 in macrophages to temper nitric oxide (NO) production and block T cell proliferation, inhibition of oxygen-requiring NO production in hypoxic regions, and induction of tryptophan-degrading enzymes that modify T cell proliferation and function. The spatial and time-dependent organization of granulomas further influences immunometabolism, for example through lactate production by activated macrophages, which can induce arginase-1. Although complex, the metabolic changes in and around TB granulomas can be potentially modified by host-directed therapies. While elimination of the TB bacilli is often the goal of any anti-TB therapy, host-directed approaches must also account for the possibility of immunopathologic damage to the lung.


Subject(s)
Energy Metabolism , Granuloma/etiology , Granuloma/metabolism , Host-Pathogen Interactions , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/pathology , Adaptation, Physiological , Amino Acids/metabolism , Animals , Arginine/metabolism , Cell Respiration , Collagen/metabolism , Host-Pathogen Interactions/immunology , Humans , Hypoxia/immunology , Hypoxia/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Nitric Oxide/biosynthesis , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Stress, Physiological , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tryptophan/metabolism , Tuberculosis/complications , Tuberculosis/microbiology
17.
Cell Rep ; 12(11): 1902-14, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26365184

ABSTRACT

Cancer can involve non-resolving, persistent inflammation where varying numbers of tumor-associated macrophages (TAMs) infiltrate and adopt different activation states between anti-tumor M1 and pro-tumor M2 phenotypes. Here, we resolve a cascade causing differential macrophage phenotypes in the tumor microenvironment. Reduction in TNF mRNA production or loss of type I TNF receptor signaling resulted in a striking pattern of enhanced M2 mRNA expression. M2 gene expression was driven in part by IL-13 from eosinophils co-recruited with inflammatory monocytes, a pathway that was suppressed by TNF. Our data define regulatory nodes within the tumor microenvironment that balance M1 and M2 populations. Our results show macrophage polarization in cancer is dynamic and dependent on the balance between TNF and IL-13, thus providing a strategy for manipulating TAMs.


Subject(s)
Macrophages/metabolism , Neoplasms/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line, Tumor , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment
18.
J Immunol ; 195(7): 3293-300, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26311904

ABSTRACT

Microbicidal NO production is reliant on inducible NO synthase-mediated L-arginine metabolism in macrophages (MΦs). However, L-arginine supply can be restricted by arginase activity, resulting in inefficient NO output and inhibition of antimicrobial MΦ function. MΦs circumvent this by converting L-citrulline to L-arginine, thereby resupplying substrate for NO production. In this article, we define the metabolic signature of mycobacteria-infected murine MΦs supplied L-arginine, L-citrulline, or both amino acids. Using liquid chromatography-tandem mass spectrometry, we determined that L-arginine synthesized from L-citrulline was less effective as a substrate for arginase-mediated L-ornithine production compared with L-arginine directly imported from the extracellular milieu. Following Mycobacterium bovis bacillus Calmette-Guérin infection and costimulation with IFN-γ, we observed that MΦ arginase activity did not inhibit production of NO derived from L-citrulline, contrary to NO inhibition witnessed when MΦs were cultured in L-arginine. Furthermore, we found that arginase-expressing MΦs preferred L-citrulline over L-arginine for the promotion of antimycobacterial activity. We expect that defining the consequences of L-citrulline metabolism in MΦs will provide novel approaches for enhancing immunity, especially in the context of mycobacterial disease.


Subject(s)
Arginine/metabolism , Citrulline/metabolism , Macrophages/metabolism , Nitric Oxide/metabolism , Tuberculosis/immunology , Animals , Arginase/metabolism , Arginine/biosynthesis , Cells, Cultured , Interferon-gamma/pharmacology , Macrophage Activation/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mycobacterium bovis/immunology , Nitric Oxide Synthase Type II/metabolism , Tuberculosis/microbiology
19.
Cancer Res ; 75(15): 3054-64, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26183929

ABSTRACT

Tristetraprolin (TTP) is an inducible zinc finger AU-rich RNA-binding protein essential for enforcing degradation of mRNAs encoding inflammatory chemokines and cytokines. Most studies on TTP center on the connection between mRNA half-life and inflammatory output, because loss of TTP amplifies inflammation by increasing the stability of AU-rich mRNAs. Here, we focused on how TTP controls cytokine and chemokine production in the nonresolving inflammation of cancer using tissue-specific approaches. In contrast with model in vitro macrophage systems, we found constitutive TTP expression in late-stage tumor-associated macrophages (TAM). However, TTP's effects on AU-rich mRNA stability were negligible and limited by constitutive p38α MAPK activity, which was the main driver of proinflammatory cytokine production in TAMs at the posttranscriptional level. Instead, elimination of TTP caused excessive protein production of inflammatory mediators, suggesting TTP-dependent translational suppression of AU-rich mRNAs. Manipulation of the p38α-TTP axis in macrophages has significant effects on the growth of tumors and therefore represents a means to manipulate inflammation in the tumor microenvironment.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Macrophages/metabolism , RNA Stability , Tristetraprolin/metabolism , Animals , Inflammation/pathology , Inflammation Mediators/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 14/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA Processing, Post-Transcriptional , Tristetraprolin/genetics
20.
Immunity ; 41(6): 947-59, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25500368

ABSTRACT

Nonresolving inflammation expands a heterogeneous population of myeloid suppressor cells capable of inhibiting T cell function. This heterogeneity has confounded the functional dissection of individual myeloid subpopulations and presents an obstacle for antitumor immunity and immunotherapy. Using genetic manipulation of cell death pathways, we found the monocytic suppressor-cell subset, but not the granulocytic subset, requires continuous c-FLIP expression to prevent caspase-8-dependent, RIPK3-independent cell death. Development of the granulocyte subset requires MCL-1-mediated control of the intrinsic mitochondrial death pathway. Monocytic suppressors tolerate the absence of MCL-1 provided cytokines increase expression of the MCL-1-related protein A1. Monocytic suppressors mediate T cell suppression, whereas their granulocytic counterparts lack suppressive function. The loss of the granulocytic subset via conditional MCL-1 deletion did not alter tumor incidence implicating the monocytic compartment as the functionally immunosuppressive subset in vivo. Thus, death pathway modulation defines the development, survival, and function of myeloid suppressor cells.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Granulocytes/physiology , Monocytes/physiology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cells/physiology , Neoplasms, Experimental/immunology , Animals , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/genetics , Caspase 8/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage/genetics , Coculture Techniques , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...