Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(5): e1012148, 2024 May.
Article in English | MEDLINE | ID: mdl-38728367

ABSTRACT

Previously, we found that Mycobacterium tuberculosis (Mtb) infection in type 2 diabetes mellitus (T2DM) mice enhances inflammatory cytokine production which drives pathological immune responses and mortality. In the current study, using a T2DM Mtb infection mice model, we determined the mechanisms that make T2DM mice alveolar macrophages (AMs) more inflammatory upon Mtb infection. Among various cell death pathways, necroptosis is a major pathway involved in inflammatory cytokine production by T2DM mice AMs. Anti-TNFR1 antibody treatment of Mtb-infected AMs from T2DM mice significantly reduced expression of receptor interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) (necroptosis markers) and IL-6 production. Metabolic profile comparison of Mtb-infected AMs from T2DM mice and Mtb-infected AMs of nondiabetic control mice indicated that 2-ketohexanoic acid and deoxyadenosine monophosphate were significantly abundant, and acetylcholine and pyridoxine (Vitamin B6) were significantly less abundant in T2DM mice AMs infected with Mtb. 2-Ketohexanoic acid enhanced expression of TNFR1, RIPK3, MLKL and inflammatory cytokine production in the lungs of Mtb-infected nondiabetic mice. In contrast, pyridoxine inhibited RIPK3, MLKL and enhanced expression of Caspase 3 (apoptosis marker) in the lungs of Mtb-infected T2DM mice. Our findings demonstrate that metabolic changes in Mtb-infected T2DM mice enhance TNFR1-mediated necroptosis of AMs, which leads to excess inflammation and lung pathology.


Subject(s)
Diabetes Mellitus, Type 2 , Mycobacterium tuberculosis , Necroptosis , Animals , Mice , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Mice, Inbred C57BL , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/microbiology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Male , Cytokines/metabolism
2.
JCI Insight ; 7(22)2022 11 22.
Article in English | MEDLINE | ID: mdl-36509283

ABSTRACT

To determine the mechanisms that mediate resistance to Mycobacterium tuberculosis (M. tuberculosis) infection in household contacts (HHCs) of patients with tuberculosis (TB), we followed 452 latent TB infection-negative (LTBI-) HHCs for 2 years. Those who remained LTBI- throughout the study were identified as nonconverters. At baseline, nonconverters had a higher percentage of CD14+ and CD3-CD56+CD27+CCR7+ memory-like natural killer (NK) cells. Using a whole-transcriptome and metabolomic approach, we identified deoxycorticosterone acetate as a metabolite with elevated concentrations in the plasma of nonconverters, and further studies showed that this metabolite enhanced glycolytic ATP flux in macrophages and restricted M. tuberculosis growth by enhancing antimicrobial peptide production through the expression of the surface receptor sialic acid binding Ig-like lectin-14. Another metabolite, 4-hydroxypyridine, from the plasma of nonconverters significantly enhanced the expansion of memory-like NK cells. Our findings demonstrate that increased levels of specific metabolites can regulate innate resistance against M. tuberculosis infection in HHCs of patients with TB who never develop LTBI or active TB.


Subject(s)
Latent Tuberculosis , Mycobacterium tuberculosis , Tuberculosis , Humans , Killer Cells, Natural
4.
JCI Insight ; 6(13)2021 07 08.
Article in English | MEDLINE | ID: mdl-34236051

ABSTRACT

In the current study, we followed 839 household contacts (HHCs) of tuberculosis (TB) patients for 2 years and identified the factors that enhanced the development of TB. Fourteen of the 17 HHCs who progressed to TB were in the 15- to 30-year-old age group. At baseline (the "0" time point, when all the individuals were healthy), the concentration of the thyroid hormone thyroxine (T4) was lower, and there were increased numbers of Tregs in PBMCs of TB progressors. At baseline, PBMCs from TB progressors stimulated with early secretory antigenic target 6 (ESAT-6) and 10 kDa culture filtrate antigen (CFP-10) produced less IL-1α. Thyroid hormones inhibited Mycobacterium tuberculosis (Mtb) growth in macrophages in an IL-1α-dependent manner. Mtb-infected Thra1PV/+ (mutant thyroid hormone receptor) mice had increased mortality and reduced IL-1α production. Our findings suggest that young HHCs who exhibit decreased production of thyroid hormones are at high risk of developing active TB disease.


Subject(s)
Leukocytes, Mononuclear/immunology , Mycobacterium tuberculosis , T-Lymphocytes, Regulatory/immunology , Thyroxine , Adolescent , Adult , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Disease Progression , Disease Transmission, Infectious/statistics & numerical data , Family Characteristics , Female , Humans , Interleukin-1alpha/metabolism , Male , Mice , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/isolation & purification , Protective Factors , Thyroxine/biosynthesis , Thyroxine/blood , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
6.
Nat Commun ; 11(1): 3535, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32669568

ABSTRACT

Macrophages are professional phagocytes known to play a vital role in controlling Mycobacterium tuberculosis (Mtb) infection and disease progression. Here we compare Mtb growth in mouse alveolar (AMs), peritoneal (PMs), and liver (Kupffer cells; KCs) macrophages and in bone marrow-derived monocytes (BDMs). KCs restrict Mtb growth more efficiently than all other macrophages and monocytes despite equivalent infections through enhanced autophagy. A metabolomics comparison of Mtb-infected macrophages indicates that ornithine and imidazole are two top-scoring metabolites in Mtb-infected KCs and that acetylcholine is the top-scoring in Mtb-infected AMs. Ornithine, imidazole and atropine (acetylcholine inhibitor) inhibit Mtb growth in AMs. Ornithine enhances AMPK mediated autophagy whereas imidazole directly kills Mtb by reducing cytochrome P450 activity. Intranasal delivery of ornithine or imidazole or the two together restricts Mtb growth. Our study demonstrates that the metabolic differences between Mtb-infected AMs and KCs lead to differences in the restriction of Mtb growth.


Subject(s)
Autophagy/drug effects , Ornithine/pharmacology , Tuberculosis/drug therapy , Urea/chemistry , Ammonia/chemistry , Animals , Apoptosis , Arginase/chemistry , Atropine/pharmacology , Cell Proliferation , Disease Progression , Female , Imidazoles/pharmacology , Kupffer Cells/drug effects , Kupffer Cells/microbiology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/microbiology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Nitric Oxide/chemistry , Phosphatidylserines/chemistry , RNA, Small Interfering/metabolism , Reactive Oxygen Species/chemistry
7.
JCI Insight ; 5(5)2020 03 12.
Article in English | MEDLINE | ID: mdl-32161191

ABSTRACT

Diabetes is a significant risk factor for the development of active tuberculosis. In this study, we used a mouse model of type 2 diabetes mellitus (T2DM) to determine the effect of prior Bacillus Calmette-Guérin (BCG) vaccination on immune responses to Mycobacterium tuberculosis (Mtb) infection. We found that, at 6-7 months after Mtb infection, 90% of the Mtb-infected T2DM mice died, whereas only 50% of BCG-vaccinated T2DM-Mtb-infected mice died. Moreover, 40% of the PBS-treated uninfected T2DM mice and 30% of the uninfected BCG-vaccinated T2DM mice died, whereas all uninfected and infected nondiabetic mice survived. BCG vaccination was less effective in reducing the lung bacterial burden of Mtb-infected T2DM mice compared with Mtb-infected nondiabetic mice. BCG vaccination significantly reduced lung inflammation in Mtb-infected T2DM mice compared with that of unvaccinated T2DM mice infected with Mtb. Furthermore, reduced mortality of BCG-vaccinated Mtb-infected T2DM mice is associated with expansion of IL-13-producing CXCR3+ Tregs in the lungs of Mtb-infected T2DM mice. Recombinant IL-13 and Tregs from BCG-vaccinated Mtb-infected T2DM mice converted proinflammatory M1 macrophages to antiinflammatory M2 macrophages. Our findings suggest a potentially novel role for BCG in preventing excess inflammation and mortality in T2DM mice infected with Mtb.


Subject(s)
BCG Vaccine/administration & dosage , Diabetes Mellitus, Type 2/complications , Tuberculosis/mortality , Animals , BCG Vaccine/immunology , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Tuberculosis/complications , Tuberculosis/immunology , Tuberculosis/prevention & control
8.
Antioxid Redox Signal ; 32(18): 1293-1312, 2020 06.
Article in English | MEDLINE | ID: mdl-32064894

ABSTRACT

Aims: Redox homeostasis is tightly controlled and regulates key cellular signaling pathways. The cell's antioxidant response provides a natural defense against oxidative stress, but excessive antioxidant generation leads to reductive stress (RS). This study elucidated how chronic RS, caused by constitutive activation of nuclear erythroid related factor-2 (caNrf2)-dependent antioxidant system, drives pathological myocardial remodeling. Results: Upregulation of antioxidant transcripts and proteins in caNrf2-TG hearts (TGL and TGH; transgenic-low and -high) dose dependently increased glutathione (GSH) redox potential and resulted in RS, which over time caused pathological cardiac remodeling identified as hypertrophic cardiomyopathy (HCM) with abnormally increased ejection fraction and diastolic dysfunction in TGH mice at 6 months of age. While the TGH mice exhibited 60% mortality at 18 months of age, the rate of survival in TGL was comparable with nontransgenic (NTG) littermates. Moreover, TGH mice had severe cardiac remodeling at ∼6 months of age, while TGL mice did not develop comparable phenotypes until 15 months, suggesting that even moderate RS may lead to irreversible damages of the heart over time. Pharmacologically blocking GSH biosynthesis using BSO (l-buthionine-SR-sulfoximine) at an early age (∼1.5 months) prevented RS and rescued the TGH mice from pathological cardiac remodeling. Here we demonstrate that chronic RS causes pathological cardiomyopathy with diastolic dysfunction in mice due to sustained activation of antioxidant signaling. Innovation and Conclusion: Our findings demonstrate that chronic RS is intolerable and adequate to induce heart failure (HF). Antioxidant-based therapeutic approaches for human HF should consider a thorough evaluation of redox state before the treatment.


Subject(s)
Antioxidants/metabolism , Cardiomyopathy, Hypertrophic/metabolism , NF-E2 Transcription Factor, p45 Subunit/metabolism , Ventricular Dysfunction, Left/metabolism , Animals , Cardiomyopathy, Hypertrophic/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Oxidative Stress , Ventricular Dysfunction, Left/pathology
9.
PLoS Pathog ; 15(12): e1008140, 2019 12.
Article in English | MEDLINE | ID: mdl-31809521

ABSTRACT

Previously, we found that pathological immune responses enhance the mortality rate of Mycobacterium tuberculosis (Mtb)-infected mice with type 2 diabetes mellitus (T2DM). In the current study, we evaluated the role of the cytokine IL-22 (known to play a protective role in bacterial infections) and type 3 innate lymphoid cells (ILC3s) in regulating inflammation and mortality in Mtb-infected T2DM mice. IL-22 levels were significantly lower in Mtb-infected T2DM mice than in nondiabetic Mtb-infected mice. Similarly, serum IL-22 levels were significantly lower in tuberculosis (TB) patients with T2DM than in TB patients without T2DM. ILC3s were an important source of IL-22 in mice infected with Mtb, and recombinant IL-22 treatment or adoptive transfer of ILC3s prolonged the survival of Mtb-infected T2DM mice. Recombinant IL-22 treatment reduced serum insulin levels and improved lipid metabolism. Recombinant IL-22 treatment or ILC3 transfer prevented neutrophil accumulation near alveoli, inhibited neutrophil elastase 2 (ELA2) production and prevented epithelial cell damage, identifying a novel mechanism for IL-22 and ILC3-mediated inhibition of inflammation in T2DM mice infected with an intracellular pathogen. Our findings suggest that the IL-22 pathway may be a novel target for therapeutic intervention in T2DM patients with active TB disease.


Subject(s)
Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/microbiology , Interleukins/immunology , Lymphocytes/immunology , Tuberculosis/immunology , Animals , Diabetes Mellitus, Type 2/complications , Humans , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Tuberculosis/complications , Interleukin-22
10.
PLoS Pathog ; 14(8): e1007174, 2018 08.
Article in English | MEDLINE | ID: mdl-30071107

ABSTRACT

In the current study, we used a mouse model and human blood samples to determine the effects of chronic alcohol consumption on immune responses during Mycobacterium tuberculosis (Mtb) infection. Alcohol increased the mortality of young mice but not old mice with Mtb infection. CD11b+Ly6G+ cells are the major source of IFN-α in the lungs of Mtb-infected alcohol-fed young mice, and IFN-α enhances macrophage necroptosis in the lungs. Treatment with an anti-IFNAR-1 antibody enhanced the survival of Mtb-infected alcohol-fed young mice. In response to Mtb, peripheral blood mononuclear cells (PBMCs) from alcoholic young healthy individuals with latent tuberculosis infection (LTBI) produced significantly higher amounts of IFN-α than those from non-alcoholic young healthy LTBI+ individuals and alcoholic and non-alcoholic old healthy LTBI+ individuals. Our study demonstrates that alcohol enhances IFN-α production by CD11b+Ly6G+ cells in the lungs of young Mtb-infected mice, which leads to macrophage necroptosis and increased mortality. Our findings also suggest that young alcoholic LTBI+ individuals have a higher risk of developing active TB infection.


Subject(s)
Alcohol Drinking/immunology , Interferon-alpha/biosynthesis , Interferon-alpha/drug effects , Tuberculosis/immunology , Adult , Animals , Disease Susceptibility/immunology , Female , Humans , Interferon-alpha/immunology , Latent Tuberculosis/immunology , Male , Mice , Mycobacterium tuberculosis
11.
Sci Rep ; 8(1): 3310, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29459675

ABSTRACT

CD4+CD25+FoxP3+ cells (Tregs) inhibit inflammatory immune responses to allografts. Here, we found that co-transplantation of allogeneic pancreatic islets with Tregs that are defective in c-Jun N-terminal kinase 1 (JNK1) signaling prolongs islet allograft survival in the liver parenchyma of chemically induced diabetic mice (CDM). Adoptively transferred JNK1-/- but not wild-type (WT) Tregs survive longer in the liver parenchyma of CDM. JNK1-/- Tregs are resistant to apoptosis and express anti-apoptotic molecules. JNK1-/- Tregs express higher levels of lymphocyte activation gene-3 molecule (LAG-3) on their surface and produce higher amounts of the anti-inflammatory cytokine interleukin (IL)-10 compared with WT Tregs. JNK1-/- Tregs inhibit liver alloimmune responses more efficiently than WT Tregs. JNK1-/- but not WT Tregs are able to inhibit IL-17 and IL-21 production through enhanced LAG-3 expression and IL-10 production. Our study identifies a novel role of JNK1 signaling in Tregs that enhances islet allograft survival in the liver parenchyma of CDM.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Graft Survival/immunology , Mitogen-Activated Protein Kinase 8/genetics , Transplantation Tolerance/immunology , Allografts/immunology , Allografts/transplantation , Animals , Antigens, CD/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Forkhead Transcription Factors/genetics , Gene Expression Regulation/immunology , Graft Survival/genetics , Humans , Interleukin-17/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukins/genetics , Mice , Mice, Inbred NOD , Mitogen-Activated Protein Kinase 8/immunology , T-Lymphocytes, Regulatory/immunology , Lymphocyte Activation Gene 3 Protein
12.
J Infect Dis ; 217(8): 1323-1333, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29390153

ABSTRACT

Background: In the current study, we determined the effects of interleukin (IL)-21 on human natural killer (NK) cells and monocyte responses during Mycobacterium tuberculosis (Mtb) infection. Methods: We found that Mtb stimulated CD4+ and NK T cells from healthy individuals with latent tuberculosis infection (LTBI+) are major sources of IL-21. CD4+ cells from tuberculosis patients secreted less IL-21 than did CD4+ cells from healthy LTBI+ individuals. Interleukin-21 had no direct effect on Mtb-stimulated monocytes. Results: Interleukin-21-activated NK cells produced interferon (IFN)-γ, perforin, granzyme B, and granulysin; lysed Mtb-infected monocytes; and reduced Mtb growth. Interleukin-21-activated NK cells also enhanced IL-1ß, IL-18, and CCL4/macrophage-inflammatory protein (MIP)-1ß production and reduced IL-10 production by Mtb-stimulated monocytes. Recombinant IL-21 (1) inhibited Mtb growth, (2) enhanced IFN-γ, IL-1ß, IL-18, and MIP-1ß, and (3) reduced IL-10 expression in the lungs of Mtb-infected Rag2 knockout mice. Conclusions: These findings suggest that activated T cells enhance NK cell responses to lyse Mtb-infected human monocytes and restrict Mtb growth in monocytes through IL-21 production. Interleukin-21-activated NK cells also enhance the immune response by augmenting IL-1ß, IL-18, and MIP-1ß production and reducing IL-10 production by monocytes in response to an intracellular pathogen.


Subject(s)
Interleukins/metabolism , Killer Cells, Natural/physiology , Tuberculosis, Pulmonary/microbiology , Animals , CD4-Positive T-Lymphocytes/physiology , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation/immunology , Humans , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , Leukocytes, Mononuclear/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes , Mycobacterium tuberculosis , Tuberculosis, Pulmonary/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...