Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Br J Cancer ; 130(10): 1659-1669, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38480935

ABSTRACT

BACKGROUND: Vestibular schwannomas (VSs) remain a challenge due to their anatomical location and propensity to growth. Macrophages are present in VS but their roles in VS pathogenesis remains unknown. OBJECTIVES: The objective was to assess phenotypic and functional profile of macrophages in VS with single-cell RNA sequencing (scRNAseq). METHODS: scRNAseq was carried out in three VS samples to examine characteristics of macrophages in the tumour. RT-qPCR was carried out on 10 VS samples for CD14, CD68 and CD163 and a panel of macrophage-associated molecules. RESULTS: scRNAseq revealed macrophages to be a major constituent of VS microenvironment with three distinct subclusters based on gene expression. The subclusters were also defined by expression of CD163, CD68 and IL-1ß. AREG and PLAUR were expressed in the CD68+CD163+IL-1ß+ subcluster, PLCG2 and NCKAP5 were expressed in CD68+CD163+IL-1ß- subcluster and AUTS2 and SPP1 were expressed in the CD68+CD163-IL-1ß+ subcluster. RT-qPCR showed expression of several macrophage markers in VS of which CD14, ALOX15, Interleukin-1ß, INHBA and Colony Stimulating Factor-1R were found to have a high correlation with tumour volume. CONCLUSIONS: Macrophages form an important component of VS stroma. scRNAseq reveals three distinct subsets of macrophages in the VS tissue which may have differing roles in the pathogenesis of VS.


Subject(s)
Macrophages , Neuroma, Acoustic , Sequence Analysis, RNA , Single-Cell Analysis , Humans , Neuroma, Acoustic/genetics , Neuroma, Acoustic/pathology , Neuroma, Acoustic/metabolism , Single-Cell Analysis/methods , Macrophages/metabolism , Macrophages/pathology , Tumor Microenvironment/genetics , Female , Male , Middle Aged , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism
2.
Eur J Neurol ; 28(6): 1910-1921, 2021 06.
Article in English | MEDLINE | ID: mdl-33768607

ABSTRACT

BACKGROUND: Evidence supports that neurodevelopmental diseases, such as Tourette syndrome (TS), may involve dysfunctional neural-immune crosstalk. This could lead to altered brain maturation and differences in immune and stress responses. Dendritic cells (DCs) play a major role in immunity as professional antigen-presenting cells; changes in their frequency have been observed in several autoimmune conditions. METHODS: In 18 TS patients (15 on stable pharmacological treatment, three unmedicated) and 18 age-matched healthy volunteers (HVs), we explored circulating blood-derived DCs and their relationship with clinical variables and brain metabolites, measured via proton magnetic resonance spectroscopy (1H-MRS). DC subsets, including plasmacytoid and myeloid type 1 and 2 dendritic cells (MDC1, MDC2), were studied with flow cytometry. 1H-MRS was used to measure total choline, glutamate plus glutamine, total creatine (tCr), and total N-acetylaspartate and N-acetylaspartyl-glutamate levels in frontal white matter (FWM) and the putamen. RESULTS: We did not observe differences in absolute concentrations of DC subsets or brain inflammatory metabolites between patients and HVs. However, TS patients manifesting anxiety showed a significant increase in MDC1s compared to TS patients without anxiety (p = 0.01). We also found a strong negative correlation between MDC1 frequency and tCr in the FWM of patients with TS (p = 0.0015), but not of HVs. CONCLUSION: Elevated frequencies of the MDC1 subset in TS patients manifesting anxiety may reflect a proinflammatory status, potentially facilitating altered neuro-immune crosstalk. Furthermore, the strong inverse correlation between brain tCr levels and MDC1 subset frequency in TS patients suggests a potential association between proinflammatory status and metabolic changes in sensitive brain regions.


Subject(s)
Tourette Syndrome , Brain/diagnostic imaging , Dendritic Cells , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy
3.
Blood ; 127(16): 1976-86, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26903549

ABSTRACT

Self-tolerance and immune homeostasis are orchestrated by FOXP3(+)regulatory T cells (Tregs). Recent data have revealed that upon stimulation, Tregs may exhibit plasticity toward a proinflammatory phenotype, producing interleukin 17 (IL-17) and/or interferon γ (IFN-γ). Such deregulation of Tregs may contribute to the perpetuation of inflammatory processes, including graft-versus-host disease. Thus, it is important to identify immunomodulatory factors influencing Treg stability. Platelet-derived microparticles (PMPs) are involved in hemostasis and vascular health and have recently been shown to be intimately involved in (pathogenic) immune responses. Therefore, we investigated whether PMPs have the ability to affect Treg plasticity. PMPs were cocultured with healthy donor peripheral blood-derived Tregs that were stimulated with anti-CD3/CD28 monoclonal antibodies in the presence of IL-2, IL-15, and IL-1ß. PMPs prevented the differentiation of peripheral blood-derived Tregs into IL-17- and IFN-γ-producing cells, even in the presence of the IL-17-driving proinflammatory cytokine IL-1ß. The mechanism of action by which PMPs prevent Treg plasticity consisted of rapid and selective P-selectin-dependent binding of PMPs to a CCR6(+)HLA-DR(+)memory-like Treg subset and their ability to inhibit Treg proliferation, in part through CXCR3 engagement. The findings that ~8% of Tregs in the circulation of healthy individuals are CD41(+)P-selectin(+)and that distinct binding of patient plasma PMPs to Tregs was observed support in vivo relevance. These findings open the exciting possibility that PMPs actively regulate the immune response at sites of (vascular) inflammation, where they are known to accumulate and interact with leukocytes, consolidating the (vascular) healing process.


Subject(s)
Blood Platelets/ultrastructure , Cell-Derived Microparticles/pathology , Cell-Derived Microparticles/physiology , Interleukin-17/metabolism , P-Selectin/physiology , T-Lymphocytes, Regulatory/metabolism , Adult , Blood Platelets/pathology , Cell Differentiation/immunology , Cells, Cultured , Down-Regulation/immunology , Forkhead Transcription Factors/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Lymphopoiesis/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology
4.
Circulation ; 131(8): 709-20, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25527700

ABSTRACT

BACKGROUND: The number of CD4(+)CD28(null) (CD28(null)) T cells, a unique subset of T lymphocytes with proinflammatory and cell-lytic phenotype, increases markedly in patients with acute coronary syndrome (ACS). ACS patients harboring high numbers of CD28(null) T cells have increased risk of recurrent severe acute coronary events and unfavorable prognosis. The mechanisms that govern the increase in CD28(null) T cells in ACS remain elusive. We investigated whether apoptosis pathways regulating T-cell homeostasis are perturbed in CD28(null) T cells in ACS. METHODS AND RESULTS: We found that CD28(null) T cells in ACS were resistant to apoptosis induction via Fas-ligation or ceramide. This was attributable to a dramatic reduction in proapoptotic molecules Bim, Bax, and Fas in CD28(null) T cells, whereas antiapoptotic molecules Bcl-2 and Bcl-xL were similar in CD28(null) and CD28(+) T cells. We also show that Bim is phosphorylated in CD28(null) T cells and degraded by the proteasome. Moreover, we demonstrate for the first time that proteasomal inhibition restores the apoptosis sensitivity of CD28(null) T cells in ACS. CONCLUSIONS: We show that CD28(null) T cells in ACS harbor marked defects in molecules that regulate T-cell apoptosis, which tips the balance in favor of antiapoptotic signals and endows these cells with resistance to apoptosis. We demonstrate that the inhibition of proteasomal activity allows CD28(null) T cells to regain sensitivity to apoptosis. A better understanding of the molecular switches that control the apoptosis sensitivity of CD28(null) T cells may reveal novel strategies for targeted elimination of these T cells in ACS patients.


Subject(s)
Acute Coronary Syndrome/metabolism , Acute Coronary Syndrome/pathology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , CD28 Antigens/deficiency , CD4-Positive T-Lymphocytes/pathology , Membrane Proteins/metabolism , Proteasome Endopeptidase Complex/physiology , Proto-Oncogene Proteins/metabolism , Acute Coronary Syndrome/epidemiology , Antibodies, Anti-Idiotypic/pharmacology , Apoptosis/drug effects , Bcl-2-Like Protein 11 , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Cells, Cultured , Fas-Associated Death Domain Protein/metabolism , Homeostasis/physiology , Humans , MAP Kinase Signaling System/physiology , Phenotype , Recurrence , Risk Factors , bcl-2-Associated X Protein/metabolism
5.
Immunology ; 146(2): 185-93, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26190355

ABSTRACT

Inflammation contributes to the development and perpetuation of several disorders and T lymphocytes orchestrate the inflammatory immune response. Although the role of T cells in inflammation is widely recognized, specific therapies that tackle inflammatory networks in disease are yet to be developed. CD4(+) CD28(null) T cells are a unique subset of helper T lymphocytes that recently shot back into the limelight as potential catalysts of inflammation in several inflammatory disorders such as autoimmunity, atherosclerosis and chronic viral infections. In contrast to conventional helper T cells, CD4(+) CD28(null) T cells have an inbuilt ability to release inflammatory cytokines and cytotoxic molecules that can damage tissues and amplify inflammatory pathways. It comes as no surprise that patients who have high numbers of these cells have more severe disease and poor prognosis. In this review, I provide an overview on the latest advances in the biology of CD4(+) CD28(null) T cells. Understanding the complex functions and dynamics of CD4(+) CD28(null) T cells may open new avenues for therapeutic intervention to prevent progression of inflammatory diseases.


Subject(s)
CD28 Antigens/immunology , Cytotoxicity, Immunologic , Inflammation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , CD28 Antigens/deficiency , Cell Death , Cell Proliferation , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation , Phenotype , Prognosis , Severity of Illness Index , Signal Transduction , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology
6.
Circ Res ; 110(6): 857-69, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22282196

ABSTRACT

RATIONALE: Patients with acute coronary syndrome (ACS) predisposed to recurrent coronary events have an expansion of a distinctive T-cell subset, the CD4(+)CD28(null) T cells. These cells are highly inflammatory and cytotoxic in spite of lacking the costimulatory receptor CD28, which is crucial for optimal T cell function. The mechanisms that govern CD4(+)CD28(null) T cell function are unknown. OBJECTIVE: Our aim was to investigate the expression and role of alternative costimulatory receptors in CD4(+)CD28(null) T cells in ACS. METHODS AND RESULTS: Expression of alternative costimulatory receptors (inducible costimulator, OX40, 4-1BB, cytotoxic T lymphocyte associated antigen-4, programmed death-1) was quantified in CD4(+)CD28(null) T cells from circulation of ACS and stable angina patients. Strikingly, in ACS, levels of OX40 and 4-1BB were significantly higher in circulating CD4(+)CD28(null) T cells compared to classical CD4(+)CD28(+) T lymphocytes. This was not observed in stable angina patients. Furthermore, CD4(+)CD28(null) T cells constituted an important proportion of CD4(+) T lymphocytes in human atherosclerotic plaques and exhibited high levels of OX40 and 4-1BB. In addition, the ligands for OX40 and 4-1BB were present in plaques and also expressed on monocytes in circulation. Importantly, blockade of OX40 and 4-1BB reduced the ability of CD4(+)CD28(null) T cells to produce interferon-γ and tumor necrosis factor-α and release perforin. CONCLUSIONS: Costimulatory pathways are altered in CD4(+)CD28(null) T cells in ACS. We show that the inflammatory and cytotoxic function of CD4(+)CD28(null) T cells can be inhibited by blocking OX40 and 4-1BB costimulatory receptors. Modulation of costimulatory receptors may allow specific targeting of this cell subset and may improve the survival of ACS patients.


Subject(s)
Acute Coronary Syndrome/immunology , CD4-Positive T-Lymphocytes/immunology , Receptors, OX40/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Acute Coronary Syndrome/metabolism , Aged , Aged, 80 and over , CD28 Antigens/genetics , CD28 Antigens/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Degranulation/immunology , Coronary Artery Disease/immunology , Coronary Artery Disease/metabolism , Female , Granzymes/metabolism , Humans , Ligands , Male , Middle Aged , Perforin/metabolism , Receptors, OX40/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
7.
J Biomed Biotechnol ; 2012: 464532, 2012.
Article in English | MEDLINE | ID: mdl-22235167

ABSTRACT

Atherosclerosis is a chronic inflammatory disease that is mediated by both the innate and adaptive immune responses. T lymphocytes, that together with B cells are the cellular effectors of the adaptive immune system, are currently endowed with crucial roles in the development and progression of atherosclerosis. Costimulatory receptors are a class of molecules expressed by T lymphocytes that regulate the activation of T cells and the generation of effector T-cell responses. In this review we present the roles of costimulatory receptors of the tumour necrosis factor receptor (TNFR) superfamily in atherosclerosis and discuss the implications for future therapies that could be used to specifically modulate the immune response of pathogenic T cells in this disease.


Subject(s)
Antigens, CD/immunology , Atherosclerosis/immunology , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor/immunology , T-Lymphocytes/immunology , Atherosclerosis/pathology , B-Lymphocytes/immunology , Humans , Immune System/immunology
8.
J Immunol ; 184(11): 6256-65, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20435933

ABSTRACT

The 129-derived Sle16 is a susceptibility locus for systemic autoimmunity when present on the C57BL/6 (B6) background. Genetic analysis of a (129xB6)F2 cross identified a region from the B6 chromosome 3 (Sle18) with positive linkage to antinuclear Abs. In this study, we have generated a B6 congenic strain harboring the 129 allele of Sle18 and intercrossed this line with the lupus-prone B6.129-Sle16 strain. The presence of the 129-Sle18 allele in the B6.129-Sle16Sle18 double congenic mice suppressed the development of Sle16-mediated autoantibody production and ameliorated the renal pathology. The 129-Sle18 locus rectified the B cell abnormalities detected in the B6.129-Sle16 mice, such as the reduction in the percentage of marginal zone B and B1a cells and the increased number of germinal centers. The B6.129-Sle16Sle18 spleens still displayed an increased percentage of activated T and B cells. However, in the B6.129-Sle16Sle18 strain the percentage of naive T cells was equivalent to that in B6.129-Sle18 and B6 mice and these cells showed a reduced proliferative response to anti-CD3 stimulation compared with B6.129-Sle16 T cells. There was a significant increase in the percentage of CD4(+)FoxP3(+)regulatory T cells in all congenic strains. These cells had normal regulatory function when tested in vitro. Thus, 129-Sle18 represents a novel, non-MHC lupus-suppressor locus probably operating as a functional modifier of B cells that, in combination with other factors, leads to lupus resistance. Further characterization of this locus will help to uncover the immune mechanism(s) conferring protection against lupus.


Subject(s)
Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/genetics , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , B-Lymphocytes/immunology , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Congenic , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology
9.
Eur J Immunol ; 40(6): 1758-67, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20213737

ABSTRACT

Complement activation is known to have deleterious effects on organ transplantation. On the other hand, the complement system is also known to have an important role in regulating immune responses. The balance between these two opposing effects is critical in the context of transplantation. Here, we report that female mice deficient in C1q (C1qa(-/-)) or C3 (C3(-/-)) reject male syngeneic grafts (HY incompatible) at an accelerated rate compared with WT mice. Intranasal HY peptide administration, which induces tolerance to syngeneic male grafts in WT mice, fails to induce tolerance in C1qa(-/-) or C3(-/-) mice. The rejection of the male grafts correlated with the presence of HY D(b)Uty-specific CD8(+) T cells. Consistent with this, peptide-treated C1qa(-/-) and C3(-/-) female mice rejecting male grafts exhibited more antigen-specific CD8(+)IFN-gamma(+) and CD8(+)IL-10(+) cells compared with WT females. This suggests that accumulation of IFN-gamma- and IL-10-producing T cells may play a key role in mediating the ongoing inflammatory process and graft rejection. Interestingly, within the tolerized male skin grafts of peptide-treated WT mice, IFN-gamma, C1q and C3 mRNA levels were higher compared to control female grafts. These results suggest that C1q and C3 facilitate the induction of intranasal tolerance.


Subject(s)
Complement C1q/immunology , Complement C3/immunology , Graft Rejection/immunology , H-Y Antigen/immunology , Transplantation, Homologous/immunology , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/immunology , Complement C1q/deficiency , Complement C3/deficiency , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , H-Y Antigen/administration & dosage , Interferon-gamma/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Skin Transplantation/immunology
10.
Blood ; 113(15): 3485-93, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19171874

ABSTRACT

Dendritic cells (DCs) are known to produce C1q, the initiator of the classical complement pathway. We demonstrate that murine DCs deficient in C1q (C1qa(-/-)) are poorer than wild-type (WT) DCs at eliciting the proliferation and Th1 differentiation of antigen-specific T cells. These defects result from decreased production of IL-12p70 by C1qa(-/-) DCs and impaired expression of costimulatory molecules CD80 and CD86 in response to CD40 ligation. The defective production of IL-12p70 and the reduced expression of CD80 and CD86 by C1qa(-/-) DCs were specifically mediated via CD40 ligation, as normal levels of IL-12p70 and CD80/86 were observed after ligation of Toll-like receptors (TLRs) on C1qa(-/-) DCs. CD40 ligation on C1qa(-/-) DCs, but not TLR ligation, results in decreased phosphorylation of p38 and ERK1/2 kinases. A strong colocalization of CD40 and C1q was observed by confocal microscopy upon CD40 ligation (but not TLR ligation) on DCs. Furthermore, human DCs from 2 C1q-deficient patients were found to have impaired IL-12p70 production in response to CD40L stimulation. Our novel data suggest that C1q augments the production of IL-12p70 by mouse and human DCs after CD40 triggering and plays important roles in sustaining the maturation of DCs and guiding the activation of T cells.


Subject(s)
CD40 Antigens/metabolism , Complement C1q/metabolism , Dendritic Cells/metabolism , Interferon-gamma/metabolism , Th1 Cells/cytology , Animals , Antigen Presentation/immunology , Apoptosis/immunology , Calreticulin/metabolism , Cell Communication/immunology , Cell Differentiation/immunology , Cells, Cultured , Complement C1q/genetics , Complement C1q/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Female , Humans , Interleukin-12/metabolism , Lymphocyte Transfusion , Male , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phagocytosis/immunology , Spleen/cytology , Th1 Cells/metabolism , Toll-Like Receptors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Immunol ; 182(5): 2795-807, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234174

ABSTRACT

Dendritic cells (DCs) have a central role in the development of adaptive immune responses, including antitumor immunity. Factors present in the tumor milieu can alter the maturation of DCs and inhibit their capacity to activate T cells. Using gene expression analysis, we found that human DCs increased the expression of TGF-beta1 transcripts following culture with human lung carcinoma cells (LCCs). These DCs produced increased amounts of TGF-beta1 protein compared with DCs not exposed to tumor cells. LCCs also decreased the expression of CD86 and HLA-DR by immature DCs. Furthermore, LCCs decreased CD86 expression and the production of TNF-alpha and IL-12 p70 by mature DCs. Moreover, LCCs also converted mature DCs into cells producing TGF-beta1. These TGF-beta1-producing DCs were poor at eliciting the activation of naive CD4(+) T cells and sustaining their proliferation and differentiation into Th1 (IFN-gamma(+)) effectors. Instead, TGF-beta1-producing DCs demonstrated an increased ability to generate CD4(+)CD25(+)Foxp3(+) regulatory T cells that suppress the proliferation of T lymphocytes. These results identify a novel mechanism by which the function of human DCs is altered by tumor cells and contributes to the evasion of the immune response.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , B7-2 Antigen/biosynthesis , B7-2 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Dendritic Cells/pathology , Down-Regulation/immunology , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/metabolism , Humans , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Lung Neoplasms/metabolism , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
12.
Cardiovasc Res ; 117(8): 1935-1948, 2021 07 07.
Article in English | MEDLINE | ID: mdl-32647892

ABSTRACT

AIMS: Inflammation has important roles in atherosclerosis. CD4+CD28null (CD28null) T cells are a specialized T lymphocyte subset that produce inflammatory cytokines and cytotoxic molecules. CD28null T cells expand preferentially in patients with acute coronary syndrome (ACS) rather than stable angina and are barely detectable in healthy subjects. Importantly, ACS patients with CD28null T-cell expansion have increased risk for recurrent acute coronary events and poor prognosis, compared to ACS patients in whom this cell subset does not expand. The mechanisms regulating CD28null T-cell expansion in ACS remain elusive. We therefore investigated the role of cytokines in CD28null T-cell expansion in ACS. METHODS AND RESULTS: High-purity sorted CD4+ T cells from ACS patients were treated with a panel of cytokines (TNF-α, IL-1ß, IL-6, IL-7, and IL-15), and effects on the number, phenotype, and function of CD28null T cells were analysed and compared to the control counterpart CD28+ T-cell subset. IL-7- and IL-15-induced expansion of CD28null T cells from ACS patients, while inflammatory cytokines TNF-α, IL-1ß, and IL-6 did not. The mechanisms underlying CD28null T-cell expansion by IL-7/IL-15 were preferential activation and proliferation of CD28null T cells compared to control CD28+ T cells. Additionally, IL-7/IL-15 markedly augmented CD28null T-cell cytotoxic function and interferon-γ production. Further mechanistic analyses revealed differences in baseline expression of component chains of IL-7/IL-15 receptors (CD127 and CD122) and increased baseline STAT5 phosphorylation in CD28null T cells from ACS patients compared to the control CD28+ T-cell subset. Notably, we demonstrate that CD28null T-cell expansion was significantly inhibited by Tofacitinib, a selective JAK1/JAK3 inhibitor that blocks IL-7/IL-15 signalling. CONCLUSION: Our novel data show that IL-7 and IL-15 drive the expansion and function of CD28null T cells from ACS patients suggesting that IL-7/IL-15 blockade may prevent expansion of these cells and improve patient outcomes.


Subject(s)
Acute Coronary Syndrome/immunology , CD28 Antigens/deficiency , CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Inflammation/immunology , Interleukin-15/pharmacology , Interleukin-7/pharmacology , Lymphocyte Activation/drug effects , Acute Coronary Syndrome/metabolism , Aged , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Female , Humans , Inflammation/metabolism , Interferon-gamma/metabolism , Janus Kinase 1/metabolism , Janus Kinase 3/metabolism , Male , Middle Aged , Phenotype , Phosphorylation , STAT5 Transcription Factor/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
13.
J Exp Med ; 200(9): 1157-65, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15504819

ABSTRACT

Strategies to enhance the immunogenicity of tumors are urgently needed. Although vaccination with irradiated dying lymphoma cells recruits a tumor-specific immune response, its efficiency as immunogen is poor. Annexin V (AxV) binds with high affinity to phosphatidylserine on the surface of apoptotic and necrotic cells and thereby impairs their uptake by macrophages. Here, we report that AxV preferentially targets irradiated lymphoma cells to CD8+ dendritic cells for in vivo clearance, elicits the release of proinflammatory cytokines and dramatically enhances the protection elicited against the tumor. The response was endowed with both memory, because protected animals rejected living lymphoma cells after 72 d, and specificity, because vaccinated animals failed to reject unrelated neoplasms. Finally, AxV-coupled irradiated cells induced the regression of growing tumors. These data indicate that endogenous adjuvants that bind to dying tumor cells can be exploited to target tumors for immune rejection.


Subject(s)
Annexin A5/immunology , Immunization , Lymphoma/immunology , Lymphoma/therapy , Receptors, Cell Surface/immunology , Ultraviolet Rays , Animals , Annexin A5/metabolism , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phagocytosis/immunology , Receptors, Cell Surface/metabolism , Tumor Cells, Cultured
14.
Blood ; 112(13): 5026-36, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18799722

ABSTRACT

Cells undergoing apoptosis are efficiently located and engulfed by phagocytes. The mechanisms by which macrophages, the professional scavenging phagocytes of apoptotic cells, are attracted to sites of apoptosis are poorly defined. Here we show that CX3CL1/fractalkine, a chemokine and intercellular adhesion molecule, is released rapidly from apoptotic lymphocytes, via caspase- and Bcl-2-regulated mechanisms, to attract macrophages. Effective chemotaxis of macrophages to apoptotic lymphocytes is dependent on macrophage fractalkine receptor, CX3CR1. CX3CR1 deficiency caused diminished recruitment of macrophages to germinal centers of lymphoid follicles, sites of high-rate B-cell apoptosis. These results provide the first demonstration of chemokine/chemokine-receptor activity in the navigation of macrophages toward apoptotic cells and identify a mechanism by which macrophage infiltration of tissues containing apoptotic lymphocytes is achieved.


Subject(s)
Apoptosis , Chemokine CX3CL1/metabolism , Chemotaxis , Lymphocytes/metabolism , Macrophages/physiology , Animals , Burkitt Lymphoma , Caspases , Cell Line, Tumor , Cells, Cultured , Humans , Lymph Nodes , Lymphocytes/cytology , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-bcl-2
15.
Front Immunol ; 10: 1644, 2019.
Article in English | MEDLINE | ID: mdl-31379843

ABSTRACT

Background: The co-inhibitory receptor PD-1 is expressed in many tumors including head and neck squamous cell carcinoma (HNSCC) and is an important immunotherapy target. However, the role of PD-1 ligands, PD-L1, and particularly PD-L2, in the tumor-stromal cell interactions that cause a tumor-permissive environment in HNSCC is not completely understood and is the focus of our study. Methods: Expression of PD-L1 and PD-L2 was analyzed by immunohistochemistry in situ in HNSCC tumor tissue. Co-cultures were established between stromal cells (fibroblasts and macrophages) and human papilloma virus (HPV)-positive and HPV-negative HNSCC cell lines (HNSCCs) and PD-1 ligands expression was analyzed using flow cytometry. Results: PD-L1 and PD-L2 were expressed both in tumor cells and stroma in HNSCC tissue in situ. In vitro, basal expression of PD-L1 and PD-L2 was low in HNSCCs and high on fibroblasts and macrophages. Interestingly, HPV-positive but not HPV-negative HNSCCs increased the expression of both PD-1 ligands on fibroblasts upon co-culture. This effect was not observed with macrophages. Conversely, both fibroblasts and macrophages increased PD-1 ligands on HPV-positive HNSCCs, whilst this was not observed in HPV-negative HNSCCs. Crucially, we demonstrate that up-regulation of PD-L1 and PD-L2 on fibroblasts by HPV-positive HNSCCs is mediated via TLR9. Conclusions: This work demonstrates in an in vitro model that HPV-positive HNSCCs regulate PD-L1/2 expression on fibroblasts via TLR9. This may open novel avenues to modulate immune checkpoint regulator PD-1 and its ligands by targeting TLR9.


Subject(s)
B7-H1 Antigen/metabolism , Head and Neck Neoplasms/metabolism , Papillomavirus Infections/metabolism , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Toll-Like Receptor 9/metabolism , Up-Regulation/physiology , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Head and Neck Neoplasms/virology , Humans , Male , Middle Aged , Papillomaviridae/pathogenicity , Papillomavirus Infections/virology , Squamous Cell Carcinoma of Head and Neck/virology , Transcriptional Activation/physiology
16.
Cardiovasc Res ; 115(9): 1385-1392, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31228191

ABSTRACT

Inflammation is an important driver of atherosclerosis, and the favourable outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial revealed the large potential of anti-inflammatory drugs for the treatment of cardiovascular disease, especially in patients with a pro-inflammatory constitution. However, the complex immune reactions driving inflammation in the vascular wall in response to an atherosclerotic microenvironment are still being unravelled. Novel insights into the cellular processes driving immunity and inflammation revealed that alterations in intracellular metabolic pathways are strong drivers of survival, growth, and function of immune cells. Therefore, this position paper presents a brief overview of the recent developments in the immunometabolism field, focusing on its role in atherosclerosis. We will also highlight the potential impact of immunometabolic markers and targets in clinical cardiovascular medicine.


Subject(s)
Arteries/immunology , Atherosclerosis/immunology , Energy Metabolism/immunology , Immune System/immunology , Immunomodulation , Inflammation/immunology , Animals , Arteries/metabolism , Arteries/physiopathology , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Consensus , Humans , Immune System/metabolism , Immune System/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Plaque, Atherosclerotic , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
J Leukoc Biol ; 81(1): 84-91, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17035340

ABSTRACT

Chemokines regulate the migration and the maturation of dendritic cells (DC) licensed by microbial constituents. We have recently found that the function of DC, including their ability to activate naïve, allogeneic CD4+ T cells, requires the autocrine/paracrine release of the nuclear protein high mobility group box 1 (HMGB1). We show here that human myeloid DC, which rapidly secrete upon maturation induction their own HMGB1, remodel their actin-based cytoskeleton, up-regulate the CCR7 and the CXCR4 chemokine receptors, and acquire the ability to migrate in response to chemokine receptor ligands. The events are apparently causally related: DC challenged with LPS in the presence of HMGB1-specific antibodies fail to up-regulate the expression of the CCR7 and CXCR4 receptors and to rearrange actin-rich structures. Moreover, DC matured in the presence of anti-HMGB1 antibodies fail to migrate in response to the CCR7 ligand CCL19 and to the CXCR4 ligand CXCL12. The blockade of receptor for advanced glycation end products (RAGE), the best-characterized membrane receptor for HMGB1, impinges as well on the up-regulation of chemokine receptors and on responsiveness to CCL19 and CXCL12. Our data suggest that the autocrine/paracrine release of HMGB1 and the integrity of the HMGB1/RAGE pathway are required for the migratory function of DC.


Subject(s)
Chemotaxis , Dendritic Cells/physiology , HMGB1 Protein/metabolism , Receptors, Immunologic/antagonists & inhibitors , Cells, Cultured , Chemokine CCL19 , Chemokine CXCL12 , Chemokines/metabolism , Chemokines, CC/metabolism , Chemokines, CXC/metabolism , Cytoskeleton/metabolism , Dendritic Cells/metabolism , Humans , Lymph Nodes/metabolism , Receptor for Advanced Glycation End Products , Receptors, CCR7 , Receptors, CXCR4/metabolism , Receptors, Chemokine/metabolism , Up-Regulation
18.
J Leukoc Biol ; 80(1): 87-95, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16617159

ABSTRACT

Pentraxins (PTX) and complement belong to the humoral arm of the innate immune system and have essential functions in immune defense to microbes and in scavenging cellular debris. The prototypic long PTX, PTX3, and the first component of the classical complement pathway, C1q, are innate opsonins involved in the disposal of dying cells by phagocytes. Whether the interaction between various innate opsonins impacts on their function is not fully understood. We show here that characterized Toll-like receptor (TLR) ligands elicit the production of C1q and PTX3 by immature dendritic cells (DC). Moreover, these molecules bind to dying cells with similar kinetics, although they recognize different domains on the cell membranes. PTX3 binds in the fluid phase to C1q, decreasing C1q deposition and subsequent complement activation on apoptotic cells. C1q increases the phagocytosis of apoptotic cells by DC and the release of interleukin-12 in the presence of TLR4 ligands and apoptotic cells; PTX3 inhibits both events. Moreover, PTX3 inhibited the cross-presentation of the MELAN-A/melanoma antigen-reactive T cell 1 (MART-1) tumor antigen expressed by dying cells, even in the presence of C1q. These results suggest that interaction of C1q and PTX3 influences the clearance of apoptotic cells by DC. The coordinated induction by primary, proinflammatory signals of C1q and PTX3 and their reciprocal regulation during inflammation influences the clearance of apoptotic cells by antigen-presenting cells and possibly plays a role in immune homeostasis.


Subject(s)
Apoptosis/immunology , C-Reactive Protein/immunology , Complement Activation/immunology , Complement C1q/immunology , Dendritic Cells/immunology , Phagocytes/immunology , Serum Amyloid P-Component/immunology , Antigen Presentation/immunology , Antigens, Neoplasm/biosynthesis , C-Reactive Protein/biosynthesis , Complement C1q/biosynthesis , Humans , MART-1 Antigen , Neoplasm Proteins/biosynthesis , Phagocytosis/immunology , Serum Amyloid P-Component/biosynthesis
19.
Article in English | MEDLINE | ID: mdl-27418972

ABSTRACT

More than 150 years from the initial description of inflammation in atherosclerotic plaques, randomized clinical trials to test anti-inflammatory therapies in atherosclerosis have recently been initiated. Lymphocytes and macrophages are main participants in the inflammatory response in atherosclerosis. T lymphocytes operate mainly by exerting strong influences on the function of many cells in the immune system and beyond, and co-ordinating their interactions. Importantly, T lymphocytes are not a homogenous population, but include several subsets with specialized functions that can either promote or suppress inflammation. The interactions between these T-lymphocyte subsets have critical consequences on the course and outcome of inflammation. The complexity of the inflammatory response in atherosclerosis poses significant challenges on translating experimental findings into clinical therapies and makes the journey from bench to bedside an arduous one. Here, we summarize recent advances on the role of CD4(+) T cells in the inflammatory process in atherosclerosis and discuss potential therapies to modulate these lymphocytes that may provide future breakthroughs in the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , T-Lymphocytes/drug effects , Animals , Atherosclerosis/blood , CD4-Positive T-Lymphocytes/drug effects , Humans , T-Lymphocytes, Regulatory/drug effects
20.
Curr Biol ; 25(5): 577-88, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25702581

ABSTRACT

BACKGROUND: Cells undergoing apoptosis are known to modulate their tissue microenvironments. By acting on phagocytes, notably macrophages, apoptotic cells inhibit immunological and inflammatory responses and promote trophic signaling pathways. Paradoxically, because of their potential to cause death of tumor cells and thereby militate against malignant disease progression, both apoptosis and tumor-associated macrophages (TAMs) are often associated with poor prognosis in cancer. We hypothesized that, in progression of malignant disease, constitutive loss of a fraction of the tumor cell population through apoptosis could yield tumor-promoting effects. RESULTS: Here, we demonstrate that apoptotic tumor cells promote coordinated tumor growth, angiogenesis, and accumulation of TAMs in aggressive B cell lymphomas. Through unbiased "in situ transcriptomics" analysis-gene expression profiling of laser-captured TAMs to establish their activation signature in situ-we show that these cells are activated to signal via multiple tumor-promoting reparatory, trophic, angiogenic, tissue remodeling, and anti-inflammatory pathways. Our results also suggest that apoptotic lymphoma cells help drive this signature. Furthermore, we demonstrate that, upon induction of apoptosis, lymphoma cells not only activate expression of the tumor-promoting matrix metalloproteinases MMP2 and MMP12 in macrophages but also express and process these MMPs directly. Finally, using a model of malignant melanoma, we show that the oncogenic potential of apoptotic tumor cells extends beyond lymphoma. CONCLUSIONS: In addition to its profound tumor-suppressive role, apoptosis can potentiate cancer progression. These results have important implications for understanding the fundamental biology of cell death, its roles in malignant disease, and the broader consequences of apoptosis-inducing anti-cancer therapy.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Neoplastic/physiology , Lymphoma, B-Cell/physiopathology , Phagocytes/physiology , Signal Transduction/physiology , Tumor Microenvironment/physiology , Analysis of Variance , Cell Proliferation/physiology , Fluorescence , Gene Expression Profiling , Histological Techniques , Humans , Kaplan-Meier Estimate , Macrophages/physiology , Matrix Metalloproteinases/metabolism , Melanoma, Experimental/physiopathology , Neovascularization, Pathologic/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL