Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 101: 194-210, 2022 03.
Article in English | MEDLINE | ID: mdl-35032575

ABSTRACT

BACKGROUND: Antigen-specific neuroinflammation and neurodegeneration are characteristic for neuroimmunological diseases. In Parkinson's disease (PD) pathogenesis, α-synuclein is a known culprit. Evidence for α-synuclein-specific T cell responses was recently obtained in PD. Still, a causative link between these α-synuclein responses and dopaminergic neurodegeneration had been lacking. We thus addressed the functional relevance of α-synuclein-specific immune responses in PD in a mouse model. METHODS: We utilized a mouse model of PD in which an Adeno-associated Vector 1/2 serotype (AAV1/2) expressing human mutated A53T-α-Synuclein was stereotactically injected into the substantia nigra (SN) of either wildtype C57BL/6 or Recombination-activating gene 1 (RAG1)-/- mice. Brain, spleen, and lymph node tissues from different time points following injection were then analyzed via FACS, cytokine bead assay, immunohistochemistry and RNA-sequencing to determine the role of T cells and inflammation in this model. Bone marrow transfer from either CD4+/CD8-, CD4-/CD8+, or CD4+/CD8+ (JHD-/-) mice into the RAG-1-/- mice was also employed. In addition to the in vivo studies, a newly developed A53T-α-synuclein-expressing neuronal cell culture/immune cell assay was utilized. RESULTS: AAV-based overexpression of pathogenic human A53T-α-synuclein in dopaminergic neurons of the SN stimulated T cell infiltration. RNA-sequencing of immune cells from PD mouse brains confirmed a pro-inflammatory gene profile. T cell responses were directed against A53T-α-synuclein-peptides in the vicinity of position 53 (68-78) and surrounding the pathogenically relevant S129 (120-134). T cells were required for α-synuclein-induced neurodegeneration in vivo and in vitro, while B cell deficiency did not protect from dopaminergic neurodegeneration. CONCLUSIONS: Using T cell and/or B cell deficient mice and a newly developed A53T-α-synuclein-expressing neuronal cell culture/immune cell assay, we confirmed in vivo and in vitro that pathogenic α-synuclein peptide-specific T cell responses can cause dopaminergic neurodegeneration and thereby contribute to PD-like pathology.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Disease Models, Animal , Dopamine , Dopaminergic Neurons/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parkinson Disease/pathology , RNA , Substantia Nigra/metabolism , T-Lymphocytes/metabolism , alpha-Synuclein/metabolism
2.
Methods Mol Biol ; 2236: 99-114, 2021.
Article in English | MEDLINE | ID: mdl-33237544

ABSTRACT

Myeloid-derived suppressor cells (MDSC) appear at relatively low frequencies in diseased organs such as tumors or infection sites, but accumulate systemically in the spleen. So far MDSC have been reported in humans and experimental animals such as mice, rats, and nonhuman primates. Therefore, methods to generate MDSC in large amounts in vitro can serve as an additional tool to study their biology. Here, we describe in detail the generation of murine MDSC with GM-CSF from bone marrow (BM). Both subsets of granulocytic (G-MDSC) and monocytic MDSC (M-MDSC) are generated by this cytokine. We provide panels of phenotypic markers to distinguish them from non-suppressive cells and define developmental stages of monocytes developing into M-MDSC by two subsequent steps in vitro.


Subject(s)
Biomarkers/metabolism , Cell Culture Techniques/methods , Myeloid-Derived Suppressor Cells/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunosuppression Therapy , Mice , Monocytes/cytology , Monocytes/drug effects , Myeloid-Derived Suppressor Cells/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
3.
Front Immunol ; 11: 616531, 2020.
Article in English | MEDLINE | ID: mdl-33584706

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) represent a major population controlling T cell immune responses. However, little is known about their molecular requirements for homing and T cell interaction to mediate suppression. Here, we investigated the functional role of the homing and collagen IV receptor VLA-1 (α1ß1-integrin) on in vitro GM-CSF generated murine MDSCs from wild-type (WT) and CD49a/α1-integrin (Itga1-/-) gene-deficient mice. Here, we found that effector (Teff) but not naive (Tn) CD4+ T cells express VLA-1 and monocytes further up-regulated their expression after culture in GM-CSF when they differentiated into the monocytic subset of resting MDSCs (R-MDSCs). Subsequent activation of R-MDSCs by LPS+IFN-γ (A-MDSCs) showed increased in vitro suppressor potential, which was independent of VLA-1. Surprisingly, VLA-1 deficiency did not influence A-MDSC motility or migration on collagen IV in vitro. However, interaction times of Itga1-/- A-MDSCs with Teff were shorter than with WT A-MDSCs on collagen IV but not on fibronectin substrate in vitro. After injection, A-MDSCs homed to the splenic red pulp where they co-localized with Teff and showed immediate suppression already after 6 h as shown by inhibition of T cell proliferation and induction of apoptosis. Injection of A-MDSCs from Itga1-/- mice showed equivalent homing into the spleen but a reduced suppressive effect. Interaction studies of A-MDSCs with Teff in the subcapsular red pulp with intravital two-photon microscopy revealed also here that MDSC motility and migration parameters were not altered by VLA-1 deficiency, but the interaction times with Teff were reduced. Together, our data point to a new role of VLA-1 adhesion to collagen IV as a prerequisite for extended contact times with Teff required for suppression.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Collagen/metabolism , Integrin alpha1beta1/metabolism , Myeloid-Derived Suppressor Cells/immunology , Spleen/immunology , Animals , Chemotaxis, Leukocyte/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
4.
Front Immunol ; 10: 2826, 2019.
Article in English | MEDLINE | ID: mdl-31849990

ABSTRACT

Monocytic myeloid-derived suppressor cells (M-MDSCs) and granulocytic MDSCs (G-MDSCs) have been found to be massively induced in TB patients as well in murine Mtb infection models. However, the interaction of mycobacteria with MDSCs and its role in TB infection is not well studied. Here, we investigated the role of Cav-1 for MDSCs infected with Mycobacterium bovis Bacille-Calmette-Guerín (BCG). MDSCs that were generated from murine bone marrow (MDSCs) of wild-type (WT) or Cav1-/- mice upregulated Cav-1, TLR4 and TLR2 expression after BCG infection on the cell surface. However, Cav-1 deficiency resulted in a selective defect of intracellular TLR2 levels predominantly in the M-MDSC subset. Further analysis indicated no difference in the phagocytosis of BCG by M-MDSCs from WT and Cav1-/- mice or caveosome formation, but a reduced capacity to up-regulate surface markers, to secrete various cytokines, to induce iNOS and NO production required for suppression of T cell proliferation, whereas Arg-1 was not affected. Among the signaling pathways affected by Cav-1 deficiency, we found lower phosphorylation of the p38 mitogen-activated protein kinase (MAPK). Together, our findings implicate that (i) Cav-1 is dispensable for the internalization of BCG, (ii) vesicular TLR2 signaling in M-MDSCs is a major signaling pathway induced by BCG, (iii) vesicular TLR2 signals are controlled by Cav-1, (iv) vesicular TLR2/Cav-1 signaling is required for T cell suppressor functions.


Subject(s)
Caveolin 1/immunology , Mycobacterium bovis , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 2/immunology , Tuberculosis/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Caveolin 1/genetics , Cytokines/immunology , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Toll-Like Receptor 4/immunology
5.
JCI Insight ; 52019 06 04.
Article in English | MEDLINE | ID: mdl-31162143

ABSTRACT

Tuberculosis patients and mice infected with live Mycobacterium tuberculosis (Mtb) accumulate high numbers of myeloid-derived suppressor cells (MDSCs). Here, we hypothesized that also dead Mtb vaccines may induce MDSCs that could impair the efficacy of vaccination. We found that repeated injections of Mtb vaccines (heat-killed Mtb in Incomplete Freund's Adjuvant, like Montanide) but not single or control vaccines without Mtb strongly expanded CD11b+ myeloid cells in the spleen, that suppressed T cell proliferation and killing ex vivo. Dead Mtb vaccination induced the generation of CD11b+ Ly-6Chigh CD115+ iNOS/Nos2+ monocytic MDSCs (M-MDSCs) upon application of inflammatory or microbial activation signals. In vivo these M-MDSCs positioned strategically in the spleen by infiltrating the splenic bridging channels and white pulp areas. Notably, within 6 to 24 hours in a Nos2-dependent fashion they produced NO to rapidly kill conventional and plasmacytoid dendritic cells (cDCs, pDCs) while, surprisingly, sparing T cells in vivo. Thus, we demonstrate that Mtb vaccine induced M-MDSCs to not directly suppress T cell in vivo but, instead, M-MDSCs directly target DC subpopulations thereby indirectly suppressing effector T cell responses. Collectively, we demonstrate that Mtb booster vaccines induce M-MDSCs in the spleen that can be activated to kill DCs cautioning to thoroughly investigate MDSC formation in individuals after Mtb vaccination in clinical trials.


Subject(s)
Dendritic Cells/immunology , Immunization, Secondary/methods , Mycobacterium tuberculosis/immunology , Myeloid-Derived Suppressor Cells/immunology , Spleen/immunology , Tuberculosis Vaccines/immunology , Animals , Antigens, Ly , CD11b Antigen , Cell Proliferation , Cytokines/immunology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Receptor, Macrophage Colony-Stimulating Factor , Spleen/pathology , T-Lymphocytes/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Vaccination
6.
Front Immunol ; 8: 726, 2017.
Article in English | MEDLINE | ID: mdl-28690613

ABSTRACT

Thymus-derived natural Foxp3+ CD4+ regulatory T cells (nTregs) play a key role in maintaining immune tolerance and preventing autoimmune disease. Several studies indicate that dendritic cells (DCs) are critically involved in the maintenance and proliferation of nTregs. However, the mechanisms how DCs manage to keep the peripheral pool at constant levels remain poorly understood. Here, we describe that the NF-κB/Rel family transcription factor RelB controls the frequencies of steady-state migratory DCs (ssmDCs) in peripheral lymph nodes and their numbers control peripheral nTreg homeostasis. DC-specific RelB depletion was investigated in CD11c-Cre × RelBfl/fl mice (RelBDCko), which showed normal frequencies of resident DCs in lymph nodes and spleen while the subsets of CD103- Langerin- dermal DCs (dDCs) and Langerhans cells but not CD103+ Langerin+ dDC of the ssmDCs in skin-draining lymph nodes were increased. Enhanced frequencies and proliferation rates were also observed for nTregs and a small population of CD4+ CD44high CD25low memory-like T cells (Tml). Interestingly, only the Tml but not DCs showed an increase in IL-2-producing capacity in lymph nodes of RelBDCko mice. Blocking of IL-2 in vivo reduced the frequency of nTregs but increased the Tml frequencies, followed by a recovery of nTregs. Taken together, by employing RelBDCko mice with increased frequencies of ssmDCs our data indicate a critical role for specific ssmDC subsets for the peripheral nTreg and IL-2+ Tml frequencies during homeostasis.

7.
Blood Adv ; 1(14): 947-960, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-29296736

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) controls proliferation and survival of myeloid cells including monocytes. Here, we describe a time-dependent licensing process driven by GM-CSF in murine Ly6Chigh and human CD14+ monocytes that disables their inflammatory functions and promotes their conversion into suppressor cells. This 2-step licensing of monocytes requires activation of the AKT/mTOR/mTORC1 signaling cascade by GM-CSF followed by signaling through the interferon-γ receptor (IFN-γR)/interferon regulatory factor-1 (IRF-1) pathway. Only licensing-dependent adaptations in Toll-like receptor/inflammasome, IFN-γR, and phosphatidylinositol 3-kinase/AKT/mTOR signaling lead to stabilized expression of inducible nitric oxide synthase by mouse and indoleamine 2,3-dioxygenase (IDO) by human monocytes, which accounts for their suppressor activity. This study suggests various myeloid cells with characteristics similar to those described for monocytic myeloid-derived suppressor cells, Mreg, or suppressor macrophages may arise from licensed monocytes. Markers of GM-CSF-driven monocyte licensing, including p-Akt, p-mTOR, and p-S6, distinguish inflammatory monocytes from potentially suppressive monocytes in peripheral blood of patients with high-grade glioma.

8.
J Exp Med ; 213(9): 1881-900, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27526711

ABSTRACT

Donor CD4(+)Foxp3(+) regulatory T cells (T reg cells) suppress graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (HCT [allo-HCT]). Current clinical study protocols rely on the ex vivo expansion of donor T reg cells and their infusion in high numbers. In this study, we present a novel strategy for inhibiting GvHD that is based on the in vivo expansion of recipient T reg cells before allo-HCT, exploiting the crucial role of tumor necrosis factor receptor 2 (TNFR2) in T reg cell biology. Expanding radiation-resistant host T reg cells in recipient mice using a mouse TNFR2-selective agonist before allo-HCT significantly prolonged survival and reduced GvHD severity in a TNFR2- and T reg cell-dependent manner. The beneficial effects of transplanted T cells against leukemia cells and infectious pathogens remained unaffected. A corresponding human TNFR2-specific agonist expanded human T reg cells in vitro. These observations indicate the potential of our strategy to protect allo-HCT patients from acute GvHD by expanding T reg cells via selective TNFR2 activation in vivo.


Subject(s)
Graft vs Host Disease/prevention & control , Receptors, Tumor Necrosis Factor, Type II/physiology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Female , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Interleukin-2/pharmacology , Mice , Mice, Inbred Strains , Myeloid-Derived Suppressor Cells/physiology
9.
Cell Rep ; 15(5): 1062-1075, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117406

ABSTRACT

Neutralization or deletion of tumor necrosis factor (TNF) causes loss of control of intracellular pathogens in mice and humans, but the underlying mechanisms are incompletely understood. Here, we found that TNF antagonized alternative activation of macrophages and dendritic cells by IL-4. TNF inhibited IL-4-induced arginase 1 (Arg1) expression by decreasing histone acetylation, without affecting STAT6 phosphorylation and nuclear translocation. In Leishmania major-infected C57BL/6 wild-type mice, type 2 nitric oxide (NO) synthase (NOS2) was detected in inflammatory dendritic cells or macrophages, some of which co-expressed Arg1. In TNF-deficient mice, Arg1 was hyperexpressed, causing an impaired production of NO in situ. A similar phenotype was seen in L. major-infected BALB/c mice. Arg1 deletion in hematopoietic cells protected these mice from an otherwise lethal disease, although their disease-mediating T cell response (Th2, Treg) was maintained. Thus, deletion or TNF-mediated restriction of Arg1 unleashes the production of NO by NOS2, which is critical for pathogen control.


Subject(s)
Arginase/metabolism , Leishmaniasis/enzymology , Leishmaniasis/pathology , Myeloid Cells/enzymology , Nitric Oxide Synthase Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acetylation/drug effects , Animals , Biomarkers/metabolism , Cell Count , Dendritic Cells/metabolism , Histones/metabolism , Interleukin-4/metabolism , Leishmania major , Leishmaniasis/immunology , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Oxidative Phosphorylation , STAT6 Transcription Factor/metabolism , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/deficiency , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Up-Regulation/drug effects
10.
Glia ; 64(5): 792-809, 2016 May.
Article in English | MEDLINE | ID: mdl-26775238

ABSTRACT

CLN diseases are mostly fatal lysosomal storage diseases that lead to neurodegeneration in the CNS. We have previously shown that CD8+ T-lymphocytes contribute to axonal perturbation and neuron loss in the CNS of Ppt1(-/-) mice, a model of CLN1 disease. We now investigated the role of the inflammation-related cell adhesion molecule sialoadhesin (Sn) in Ppt1(-/-) and Cln3(-/-) mice, a model of the most frequent form, CLN3 disease. Microglia/macrophages in the CNS of both models showed an upregulation of Sn and markers for proinflammatory M1 polarization and antigen presentation. Sn+ microglia/macrophages associated with SMI32+ axonal spheroids and CD8+ T-lymphocytes. To analyze their pathogenic impact, we crossbred both models with Sn-deficient mice and scored axonal degeneration and neuronal integrity using immunohistochemistry, electron microscopy and optical coherence tomography. Degenerative alterations in the retinotectal pathway of Ppt1(-/-)Sn(-/-) and Cln3(-/-)Sn(-/-) mice were significantly reduced. Ppt1(-/-)Sn(-/-) mice also showed a substantially improved clinical phenotype and extended lifespan, attenuated numbers of M1-polarized microglia/macrophages and reduced expression levels of proinflammatory cytokines. This was accompanied by an increased frequency of CD8+CD122+ T-lymphocytes in the CNS of Ppt1(-/-)Sn(-/-) mice, the regulatory phenotype of which was demonstrated by impaired survival of CD8+CD122- effector T-lymphocytes in co-culture experiments. We show for the first time that increased Sn expression on microglia/macrophages contributes to neural perturbation in two distinct models of CLN disease. Our data also indicate that a rarely described CD8+CD122+ T-cell population can regulate the corresponding diseases. These studies provide insights into CLN pathogenesis and may guide in designing immuno-regulatory treatment strategies.


Subject(s)
Astrocytes/pathology , Encephalitis/etiology , Neuronal Ceroid-Lipofuscinoses/complications , Neuronal Ceroid-Lipofuscinoses/pathology , Sialic Acid Binding Ig-like Lectin 1/metabolism , Animals , Antigens, CD/metabolism , Astrocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcium-Binding Proteins/metabolism , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Disease Progression , Encephalitis/pathology , Macrophages/metabolism , Macrophages/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/metabolism , Microglia/pathology , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Retina/pathology , Retina/ultrastructure , Sialic Acid Binding Ig-like Lectin 1/genetics , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Tomography, Optical Coherence
11.
BMC Immunol ; 14: 48, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24089996

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis (Mtb) infections are still a major cause of death among all infectious diseases. Although 99% of individuals infected with Mtb develop a CD4(+) Th1 and CD8(+) T cell mediated immunity as measured by tuberculin skin test, this results only in partial protection and Mtb vaccines are not effective. Deviation of immune responses by pathogens towards a Th2 profile is a common mechanism of immune evasion, typically leading to the persistence of the microbes. RESULTS: Here we tested the stimulatory capacity of selective Mtb antigens on human monocyte-derived dendritic cell (DC) maturation and cytokine production. DC maturation markers CD80, CD86 and CD83 were readily upregulated by H37Ra- and H37Rv-associated antigens, the 30-kDa (from Ag85 B complex) and 38-KDa Mtb antigens only partially induced these markers. All Mtb antigens induced variable levels of IL-6 and low levels of IL-10, there was no release of IL-12p70 detectable. Substantial IL-12p40 production was restricted to LPS or H37Ra and H37Rv preparations. Although the proliferation levels of primary T cell responses were comparable using all the differentially stimulated DC, the 30-kDa and 38-kDa antigens showed a bias towards IL-4 secretion of polarized CD4(+) T cells after secondary stimulation as compared to H37Ra and H37Rv preparations. CONCLUSION: Together our data indicate that 30-kDa and 38-kDa Mtb antigens induced only partial DC maturation shifting immune responses towards a Th2 profile.


Subject(s)
Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interleukin-4/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/pharmacology , Antigens, CD/immunology , Antigens, CD/metabolism , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Flow Cytometry , Humans , Immunoglobulins/immunology , Immunoglobulins/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Interleukin-4/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Molecular Weight , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/immunology , CD83 Antigen
12.
Am J Respir Crit Care Med ; 188(6): 724-32, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23885784

ABSTRACT

RATIONALE: Inadequacy of T-cell responses may result in the development of tuberculosis (TB). Myeloid-derived suppressor cells (MDSCs) have been described as suppressors of T-cell function in cancer biology and recently in several infectious diseases. OBJECTIVES: To explore the presence and role of MDSCs in TB. METHODS: We analyzed surface markers of MDSCs in peripheral blood and at the site of disease in TB cases and in patients with lung cancer, and in peripheral blood of asymptomatic tuberculin skin test-positive individuals with recent (household) or remote exposure to Mycobacterium tuberculosis (M.tb) and in uninfected healthy control subjects. To evaluate the suppressive capacity of MDSCs, cells of household contacts infected with M.tb and TB cases were isolated and cocultured with CD3(+) T cells. MEASUREMENTS AND MAIN RESULTS: Our results demonstrate an increased presence of MDSCs after recent M.tb infection and disease. We confirm their suppression of CD4(+) T-cell function, including reduced cytokine responses and inhibition of CD4(+) T-cell proliferation. Only MDSCs from TB cases reduced T-cell activation, altered T-cell trafficking, and suppressed CD8(+) T-cell functions. M.tb-expanded MDSCs were associated with significantly higher IL-1ß, IL-6, IL-8, granulocyte colony-stimulating factor, and monocyte chemotactic protein-1, and reduced granulocyte-macrophage colony-stimulating factor and macrophage inflammatory protein-1 beta levels in coculture. CONCLUSIONS: These data reveal that innate MDSCs are induced not only during active TB at similar levels as found in cancer, but also in healthy individuals after recent exposure to M.tb. These cells diminish protective T-cell responses and may contribute to the inability of hosts to eradicate the infection and add to the subsequent development of TB disease.


Subject(s)
Mycobacterium Infections/immunology , Mycobacterium tuberculosis/immunology , Myeloid Cells/immunology , T-Lymphocytes/immunology , Tuberculosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Cytokines/immunology , Flow Cytometry/methods , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-1beta/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Lung Neoplasms/blood , Mycobacterium Infections/blood , Tuberculin Test/methods , Tuberculosis/blood
13.
PLoS One ; 7(11): e50080, 2012.
Article in English | MEDLINE | ID: mdl-23226238

ABSTRACT

The ability of CD4(+)Foxp3(+) regulatory T-cells (Treg) to produce interleukin (IL)-10 is important for the limitation of inflammation at environmental interfaces like colon or lung. Under steady state conditions, however, few Tregs produce IL-10 ex vivo. To investigate the origin and fate of IL-10 producing Tregs we used a superagonistic mouse anti-mouse CD28 mAb (CD28SA) for polyclonal in vivo stimulation of Tregs, which not only led to their numeric expansion but also to a dramatic increase in IL-10 production. IL-10 secreting Tregs strongly upregulated surface receptors associated with suppressive function as compared to non-producing Tregs. Furthermore, polyclonally expanding Tregs shifted their migration receptor pattern after activation from a CCR7(+)CCR5(-) lymph node-seeking to a CCR7(-)CCR5(+) inflammation-seeking phenotype, explaining the preferential recruitment of IL-10 producers to sites of ongoing immune responses. Finally, we observed that IL-10 producing Tregs from CD28SA stimulated mice were more apoptosis-prone in vitro than their IL-10 negative counterparts. These findings support a model where prolonged activation of Tregs results in terminal differentiation towards an IL-10 producing effector phenotype associated with a limited lifespan, implicating built-in termination of immunosuppression.


Subject(s)
Antibodies/pharmacology , Apoptosis/drug effects , CD28 Antigens/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/immunology , CD28 Antigens/agonists , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Clone Cells , Gene Expression/drug effects , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Receptors, CCR5/genetics , Receptors, CCR5/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects
14.
J Clin Invest ; 121(7): 2898-910, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21633167

ABSTRACT

Immune mechanisms are known to control the pathogenesis of atherosclerosis. However, the exact role of DCs, which are essential for priming of immune responses, remains elusive. We have shown here that the DC-derived chemokine CCL17 is present in advanced human and mouse atherosclerosis and that CCL17+ DCs accumulate in atherosclerotic lesions. In atherosclerosis-prone mice, Ccl17 deficiency entailed a reduction of atherosclerosis, which was dependent on Tregs. Expression of CCL17 by DCs limited the expansion of Tregs by restricting their maintenance and precipitated atherosclerosis in a mechanism conferred by T cells. Conversely, a blocking antibody specific for CCL17 expanded Tregs and reduced atheroprogression. Our data identify DC-derived CCL17 as a central regulator of Treg homeostasis, implicate DCs and their effector functions in atherogenesis, and suggest that CCL17 might be a target for vascular therapy.


Subject(s)
Atherosclerosis/immunology , Chemokine CCL17/immunology , Dendritic Cells/immunology , Homeostasis , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation , Cell Movement , Chemokine CCL17/genetics , Humans , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/metabolism
15.
Med Microbiol Immunol ; 199(3): 273-81, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20376485

ABSTRACT

Tumor cells and microorganisms manipulate the immune system to minimize any counter response in order to survive. Myeloid-derived suppressor cells (MDSC) in the mouse represent activated Gr-1(+) CD11b(+) myeloid precursor cells. Activation may occur through endogenous or exogenous factors leading to the suppression of immune responses. Under steady state conditions the same precursors differentiate into dendritic cells, macrophages and neutrophils. Their linkage to tumor progression and several suppression mechanisms employing the arginine metabolism are well documented, but knowledge of their role in chronic infections, autoimmune diseases and graft-versus-host reactions is just emerging. Several factors have been described to promote MDSC expansion and activation in bone marrow, spleen and tumor sites. New evidence suggests that the Gr-1 antibody itself may differentially trigger myelopoiesis under steady state conditions or induce apoptosis in inflammatory situations after binding to a common epitope expressed on Ly-6C and Ly-6G molecules, respectively. Moreover, two subsets of neutrophil- and monocyte-related MDSC have been described in tumor-bearing and healthy mice. In the present review, we summarize some early work leading to recent findings on these two MDSC subsets, the factors supporting MDSC expansion and activation, as well as novel insights on Gr-1 antibody functions.


Subject(s)
Myeloid Progenitor Cells/immunology , Animals , Bone Marrow/immunology , CD11b Antigen/immunology , Cell Differentiation/immunology , Cell Proliferation , Humans , Immune Tolerance , Mice , Neoplasms/immunology , Spleen/cytology , Spleen/immunology
16.
J Med Virol ; 82(3): 396-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20087929

ABSTRACT

Human herpesvirus 6 (HHV-6) is widespread in the human population by infecting most individuals in early childhood. After primary infection, HHV-6 establishes a latent infection by remaining in circulating mononuclear cells of healthy individuals. The HHV-6 antibody titer increases after primary infection with measles virus. The present study was undertaken to determine the specific antiviral IgG1, IgG2, IgG3, and IgG4 subclass response patterns to HHV-6 in HHV-6-seropositive individuals with natural measles virus infection, measles vaccination, and rubella virus infection. The purpose of this study was to examine HHV-6-specific IgG isotype response in patients with acute virus coinfection. Serum samples were obtained from individuals who were seropositive for HHV-6 after natural primary infection with measles virus during an outbreak, measles vaccination, or rubella virus infection, and from healthy individuals. Sera were examined by indirect immunofluorescence assays for detection of HHV-6-specific IgG1, IgG2, IgG3, and IgG4 antibodies. A high percentage (69%) of those infected with measles virus had an HHV-6 IgG1 and IgG4 response (P < 0.001, chi(2) test), whereas persons vaccinated against measles, those infected with rubella, and healthy individuals showed an HHV-6 IgG1 response. These results demonstrate that natural measles virus infection induces an HHV-6 IgG isotype response, which suggests a shift in immune activity from a Th1 to a Th2 response. J. Med. Virol. 82:396-399, 2010. (c) 2010 Wiley-Liss, Inc.


Subject(s)
Antibodies, Viral/blood , Herpesvirus 6, Human/immunology , Immunoglobulin G/blood , Measles Vaccine/immunology , Measles/immunology , Rubella/immunology , Adolescent , Adult , Child , Child, Preschool , Fluorescent Antibody Technique, Indirect , Humans , Infant , Middle Aged , Young Adult
17.
Eur J Immunol ; 39(12): 3538-51, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19830733

ABSTRACT

The Gr-1 (RB6-8C5) Ab binds with high affinity to mouse Ly-6G molecules and to a lower extent to Ly-6C and has been widely used for cell depletion in infected or tumor-bearing mice. Here we found that Gr-1 treatment of BM cells in vitro and in vivo showed no depleting effects. The epitope recognized by the Gr-1 Ab overlapped with Ly-6G (1A8 Ab) but not Ly-6C (ER-MP20 Ab). In vitro the Gr-1 Ab transmitted signals via STAT-1, STAT-3 and STAT-5 into BM cells, similar to GM-CSF. In healthy mice injected with the Gr-1 Ab, the Ab remained attached to the surface of myeloid cells for at least four days. Gr-1 Ab induced myeloid cell expansion, upregulation of macrophage markers, but not the DC marker CD11c. Suppressor activity of two distinct Gr-1(high) and Gr-1(low) expressing BM-myeloid-derived suppressor cell subsets was transiently ablated by Gr-1 Ab injection. Depleting effects of Gr-1 Ab could only be observed on inflammatory Ly-6C(int)Ly-6G(high) neutrophils from the peritoneal cavity, which occurred via apoptosis and was associated with the absence of Mcl-1 expression. Together, Gr-1 Ab induces signals leading to myelopoiesis and affects myeloid-derived suppressor cell activity, suggesting functional roles for Ly-6C/G molecules in macrophage differentiation and neutrophil apoptosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bone Marrow Cells/drug effects , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/metabolism , Antigens, Ly/immunology , Antigens, Ly/metabolism , Apoptosis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Macrophage-1 Antigen , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myelopoiesis/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism
18.
Eur J Immunol ; 39(10): 2865-76, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19637228

ABSTRACT

Myeloid-derived suppressor cells (MDSC) and DC are major controllers of immune responses against tumors or infections. However, it remains unclear how DC development and MDSC suppressor activity both generated from myeloid precursor cells are regulated. Here, we show that the combined treatment of BM-derived MDSC with LPS plus IFN-gamma inhibited the DC development but enhanced MDSC functions, such as NO release and T-cell suppression. This was not observed by the single treatments in vitro. In the spleens of healthy mice, we identified two Gr-1(low)CD11b(high)Ly-6C(high)SSC(low)Mo-MDSC and Gr-1(high)CD11b(low)PMN-MDSC populations with suppressive potential, whereas Gr-1(high)CD11b(high) neutrophils and Gr-1(low)CD11b(high)SSC(low) eosinophils were not suppressive. Injections of LPS plus IFN-gamma expanded these populations within the spleen but not LN leading to the block of the proliferation of CD8(+) T cells. At the same time, their capacity to develop into DC was impaired. Together, our data suggest that spleens of healthy mice contain two subsets of MDSC with suppressive potential. A two-signal-program through combined LPS and IFN-gamma treatment expands and fully activates MDSC in vitro and in vivo.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Immune Tolerance/immunology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Myeloid Cells/drug effects , Adoptive Transfer , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD11b Antigen/metabolism , Cell Differentiation/immunology , Cytokines/pharmacology , Dendritic Cells/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immune Tolerance/drug effects , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Nitric Oxide/metabolism , Ovalbumin/immunology , Receptors, Chemokine/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
19.
J Immunol ; 176(2): 923-30, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16393977

ABSTRACT

The human herpesvirus 8 (HHV-8) is a gamma herpesvirus with oncogenic potential which establishes a chronic infection that is normally controlled by the immune system of healthy individuals. In particular, CTL responses seem to play a key role in control of the infection. In this study, we characterized epitope-specific CTL responses in healthy HHV-8-seropositive individuals against four HHV-8 lytic Ags: open reading frames (ORF) 26, 70, K3, and K5. We found that the majority of subjects responded to at least one HHV-8 lytic Ag-derived epitope, and some of these epitopes represented dominant targets, suggesting that they could be relevant targets of CTL-mediated immunity in vivo, and may be involved in host control of HHV-8. Specifically, we identified three CTL epitopes from ORF 26, which are presented by HLA-A2, six CTL epitopes from ORF 70 presented by HLA-A2 (three epitopes), -A24 (two epitopes), and -B7 (one epitope), three CTL epitopes from ORF K3 presented by HLA-A2 (two epitopes) and -B7 (one epitope), and one HLA-A2 presented epitope derived from ORF K5. The identified epitopes may be regarded as useful tools for understanding the role of CTL responses to lytic Ags in individuals affected by HHV-8-associated disorders, and for the development of immunotherapies for the treatment/prevention of HHV-8-associated malignancies.


Subject(s)
Antigens, Viral , CD8-Positive T-Lymphocytes/immunology , Herpesvirus 8, Human/immunology , Amino Acid Sequence , Antigen Presentation , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cell Line , Epitopes/genetics , Epitopes/metabolism , Genes, Viral , HLA-A Antigens/metabolism , HLA-A2 Antigen/metabolism , HLA-A24 Antigen , HLA-B7 Antigen/metabolism , Herpesviridae Infections/immunology , Herpesvirus 8, Human/genetics , Humans , Immunologic Memory , In Vitro Techniques , Open Reading Frames , Protein Binding , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL