Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Immunol ; 11: 1640, 2020.
Article in English | MEDLINE | ID: mdl-32849561

ABSTRACT

Every sixth child suffers from hypertrophy of the adenoid, a secondary lymphoid organ, at least once in childhood. Little is known about the impact of pathogen-provocation vs. developmental impact on T-cell responses after 1 year of age. Therefore, developmental and infection-driven influences on the formation of T-cell-compartments and -multifunctionality in adenoids were analyzed taking into account patient's history of age and inflammatory processes. Here, we show that in adenoids of 102 infants and children similar frequencies of naïve, effector, and memory T-cells were accumulated, whereby history of suffering from subsequent infection symptoms resulted in lower frequencies of CD4+ and CD8+ T-cells co-expressing several cytokines. While patients suffering from sole nasal obstruction had balanced Th1- and Th17-compartments, Th1 dominated in patients with concomitant upper airway infections. In addition, analysis of cytokine co-expressing CD4+ and CD8+ T-cells showed that children at the age of three or older differed significantly from those being 1- or 2-years old, implicating a developmental switch in T-cell differentiation at that age. Yet, dissecting age and infectious history of the patients revealed that while CD8+ T-cell differentiation seems to be triggered by development, CD4+ T-cell functionality is partly impaired by infections. However, this functionality recovers by the age of 3 years. Thus, 3 years of age seems to be a critical period in an infant's life to develop robust T-cell compartments of higher quality. These findings identify important areas for future research and distinguish an age period in early childhood when to consider adjusting the choice of treatment of infections.


Subject(s)
Cell Differentiation/immunology , Immunity, Cellular , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adenoids/immunology , Adenoids/metabolism , Adolescent , Age Factors , Cell Differentiation/genetics , Child , Child, Preschool , Disease Susceptibility , Female , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Immunologic Memory , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Count , Male , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/cytology
2.
J Am Heart Assoc ; 3(2): e000611, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24732919

ABSTRACT

BACKGROUND: The pathogenesis of collateral growth (arteriogenesis) has been linked to both the innate and adaptive immune systems. While therapeutic approaches for the augmentation of arteriogenesis have focused on innate immunity, exploiting both innate and adaptive immune responses has not been examined. We hypothesized that tetanus toxoid (tt) immunization of mice followed by transplantation of monocytes (Mo) exposed ex vivo to tt augments arteriogenesis after ligation of the hind limb. METHODS AND RESULTS: Mo were generated from nonimmunized BALB/c mice, exposed ex vivo to tt for 24 hours and intravenously injected (ttMo, 2.5×10(6)) into the tail veins of tt-immunized syngeneic mice whose hind limbs had been ligated 24 hours prior to transplantation. Laser Doppler perfusion imaging was applied, and a perfusion index (PI) was calculated (ratio ligated/unligated). Twenty-one days after ligation, the arteriogenesis of untreated BALB/c mice was limited (PI=0.49±0.09). Hind limb function was impaired in 80% of animals. Injection of non-engineered Mo insignificantly increased the PI to 0.56±0.07. However, ttMo transplantation resulted in a strong increase of the PI to 0.82±0.08 (n=7; P<0.001), with no (0%) detectable functional impairment. ttMo injected into nonimmunized mice had no effect. The strong arteriogenic response of ttMo transplantation into immunized mice was prevented when mice had been depleted of T-helper cells by CD4-antibody pretreatment (PI=0.50±0.08; n=17; P<0.001), supporting the hypothesis that transplanted cells interact with recipient lymphocytes. CONCLUSIONS: Transplantation of ttMo into pre-immunized mice strongly promotes arteriogenesis. This therapeutic approach is feasible and highly attractive for the alleviation of morbidity associated with vascular occlusive disease.


Subject(s)
Collateral Circulation , Immunization , Ischemia/therapy , Monocytes/transplantation , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Tetanus Toxoid/administration & dosage , Adaptive Immunity , Animals , Cells, Cultured , Coculture Techniques , Cytokines/blood , Disease Models, Animal , Hindlimb , Immunity, Innate , Ischemia/blood , Ischemia/immunology , Ischemia/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Recovery of Function , Regional Blood Flow , T-Lymphocytes/immunology , Tetanus Toxoid/immunology , Time Factors
3.
J Immunol ; 192(11): 5160-70, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24778440

ABSTRACT

Senescence or biological aging impacts a vast variety of molecular and cellular processes. To date, it is unknown whether CD4(+) Th cells display an age-dependent bias for development into specific subpopulations. In this study, we show the appearance of a distinct CD4(+) T cell subset expressing IL-4 at an early stage of development in infant adenoids and cord blood that is lost during aging. We identified by flow cytometric, fluorescent microscopic, immunoblot, and mass spectrometric analysis a population of CD4(+) T cells that expressed an unglycosylated isoform of IL-4. This T cell subpopulation was found in neonatal but not in adult CD4(+) T cells. Furthermore, we show that the mRNA of the Th2 master transcription factor GATA3 is preferentially expressed in neonatal CD4(+) T cells. The Th2 phenotype of the IL-4(+)CD4(+) T cells could be reinforced in the presence of TGF-ß. Although the IL-4(+)CD4(+) T cells most likely originate from CD31(+)CD4(+) T recent thymic emigrants, CD31 was downregulated prior to secretion of IL-4. Notably, the secretion of IL-4 requires a so far unidentified trigger in neonatal T cells. This emphasizes that cytokine expression and secretion are differentially regulated processes. Our data support the hypothesis of an endogenously poised cytokine profile in neonates and suggest a link between cytokine production and the developmental stage of an organism. The determination of the IL-4 isoform-expressing cells in humans might allow the identification of Th2 precursor cells, which could provide novel intervention strategies directed against Th2-driven immunopathologies such as allergies.


Subject(s)
Interleukin-4/immunology , Th2 Cells/immunology , Female , GATA3 Transcription Factor/immunology , Gene Expression Regulation/immunology , Glycosylation , Humans , Hypersensitivity/immunology , Infant , Infant, Newborn , Male , Protein Isoforms/immunology , Th2 Cells/cytology , Transforming Growth Factor beta/immunology
5.
J Immunol ; 187(11): 5627-35, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22048775

ABSTRACT

Cytokines are critical messengers that control the differentiation of Th cells. To evaluate their impact on the fate of human naive CD4(+) T cells from cord and adult blood, early T cell differentiation was monitored after T cell activation in the presence of pro- and anti-inflammatory cytokines. Interestingly, the analysis of Th cell lineage-specific molecules revealed that IL-1ß on its own mediates differentiation of Th cells that secrete a wide range of proinflammatory cytokines and stably express CD69, STAT1, IFN-γ, and IL-17. Notably, our data suggest that IL-1ß induces Th17 cells independent of RORC upregulation. In contrast, TGF-ß that triggers RORC prevents Th17 cell development. This suppressive function of TGF-ß is characterized by inhibition of STAT1, STAT3, and CD69. However, after repeated anti-CD3 and anti-CD28 stimulation, we observe that TGF-ß provokes an increase in Th17 cells that presumably relies on reactivation of a default pathway by preferential inhibition of IFN-γ. Hence, our data extend the view that the principal cytokines for determining Th cell fate are IL-12 for the Th1 lineage, IL-4 for the Th2 lineage, and TGF-ß in conjunction with IL-6 for the Th17 lineage. We propose that IL-1ß induces a general proinflammatory Th cell precursor that, in the presence of the lineage-specifying cytokines, further differentiates into one of the specific Th cell subpopulations.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Interleukin-1beta/metabolism , Transforming Growth Factor beta/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Separation , Flow Cytometry , Humans , Immunoblotting , Interleukin-1beta/immunology , Lymphocyte Activation/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/immunology
6.
J Immunol ; 186(10): 5580-9, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21478403

ABSTRACT

CTLA-4 is known as a central inhibitor of T cell responses. It terminates T cell activation and proliferation and induces resistance against activation induced cell death. However, its impact on memory formation of adaptive immune responses is still unknown. In this study, we demonstrate that although anti-CTLA-4 mAb treatment during primary immunization of mice initially enhances the number of IFN-γ-producing CD4(+) T cells, it does not affect the size of the memory pool. Interestingly, we find that the CTLA-4 blockade modulates the quality of the memory pool: it decreases the amount of specialized "multifunctional" memory CD4(+) T cells coproducing IFN-γ, TNF-α, and IL-2 in response to Ag. The reduction of these cells causes an immense decrease of IFN-γ-producing T cells after in vivo antigenic rechallenge. Chimeric mice expressing CTLA-4-competent and -deficient cells unmask, which these CTLA-4-driven mechanisms are mediated CD4(+) T cell nonautonomously. In addition, the depletion of CD25(+) T cells prior to the generation of Ag-specific memory cells reveals that the constitutively CTLA-4-expressing natural regulatory T cells determine the quality of memory CD4(+) T cells. Taken together, these results indicate that although the inhibitory molecule CTLA-4 damps the primary immune response, its engagement positively regulates the formation of a high-quality memory pool equipped with multifunctional CD4(+) T cells capable of mounting a robust response to Ag rechallenge.


Subject(s)
Antigens, CD/immunology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocyte Activation , T-Lymphocyte Subsets/immunology , Adaptive Immunity , Animals , Antibodies, Monoclonal/immunology , Antigens/immunology , Antigens, CD/genetics , CD40 Ligand/genetics , CTLA-4 Antigen , Cells, Cultured , Chimera , Flow Cytometry , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-2 Receptor alpha Subunit , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Immunol ; 182(9): 5342-51, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380781

ABSTRACT

CD28(null) T cells are a highly enriched subset of proinflammatory T cells in patients with autoimmune diseases that are oligoclonal and autoreactive. In this study, we analyzed the role of CD152 signaling on the longevity of human CD28(null) T cells. Using a sensitive staining method for CD152, we show that human CD4(+)CD28(null) and CD8(+)CD28(null) T cells rapidly express surface CD152. Serological inactivation of CD152 using specific Fab or blockade of CD152 ligands using CTLA-4Ig in CD4(+)CD28(null) and CD8(+)CD28(null) T cells enhances apoptosis in a Fas/FasL-dependent manner. CD152 cross-linking on activated CD28(null) cells prevents activation-induced cell death as a result of reduced caspase activity. Apoptosis protection conferred by CD152 is mediated by phosphatidylinositol 3'-kinase-dependent activation of the kinase Akt, resulting in enhanced phosphorylation and thereby inhibition of the proapoptotic molecule Bad. We show that signals triggered by CD152 act directly on activated CD28(null) T lymphocytes and, due to its exclusive expression as a receptor for CD80/CD86 on CD28(null) T cells, prevention of CD152-mediated signaling is likely a target mechanism taking place during therapy with CTLA-4Ig. Our data imply strongly that antagonistic approaches using CD152 signals for chronic immune responses might be beneficial.


Subject(s)
Antigens, CD/biosynthesis , CD28 Antigens , Lymphocytes, Null/cytology , Lymphocytes, Null/immunology , Membrane Proteins/biosynthesis , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Antigens, CD/genetics , Antigens, CD/physiology , Apoptosis/immunology , CD28 Antigens/metabolism , CTLA-4 Antigen , Cell Cycle/immunology , Cell Proliferation , Cell Survival/immunology , Humans , Lymphocyte Activation/immunology , Lymphocytes, Null/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , T-Lymphocyte Subsets/metabolism
8.
Eur J Immunol ; 36(11): 2912-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17051619

ABSTRACT

Dendritic cells (DC) play an important role as antigen-presenting cells in T cell stimulation. Interestingly, a number of recent studies also imply DC as critical accessory cells in B cell activation, isotype switching and plasma blast maintenance. Here we use the conditional in vivo ablation of CD11c(high) DC to investigate the role of these cells in T-independent type 2 immune responses. We show that CD11c(high) DC are dispensable for the initiation and maintenance of a primary immune response against the T-independent type 2 antigen (4-hydroxy-3-nirophenyl)acetyl-Ficoll. Our results suggest that support for plasma cell formation in T cell-independent immune responses can be provided by non-DC such as stromal cells, or is independent of external signals. Interestingly, we found plasma blasts to express CD11c and to be diphtheria toxin-sensitive in CD11c-diphtheria toxin receptor-transgenic mice, providing a unique tool for future analysis of in vivo aspects of plasma cell biology.


Subject(s)
CD11c Antigen/metabolism , Dendritic Cells/immunology , Plasma Cells/immunology , Animals , Antibodies/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD11c Antigen/analysis , CD11c Antigen/genetics , Cell Differentiation , Dendritic Cells/chemistry , Dendritic Cells/cytology , Diphtheria Toxin/pharmacology , Ficoll/analogs & derivatives , Ficoll/immunology , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Plasma Cells/cytology , Plasma Cells/drug effects , Receptors, Cell Surface/genetics , T-Lymphocytes/immunology
9.
Immunol Rev ; 211: 295-302, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16824136

ABSTRACT

Immunological memory, as provided by antibodies, depends on the continued presence of antibody-secreting cells, such as long-lived plasma cells of the bone marrow. Survival niches for these memory plasma cells are limited in number. In an established immune system, acquisition of new plasma cells, generated in response to recent pathogenic challenges, requires elimination of old memory plasma cells. Here, we review the adaptation of plasma cell memory to new pathogens. This adaptation is dependent upon the influx of plasmablasts, generated in a secondary systemic immune reaction, into the pool of memory plasma cells, the efficiency of competition of new plasmablasts with old plasma cells, and the frequency of infection with novel pathogens. To maintain old plasma cells at frequencies high enough to provide protection and to accommodate as many specificities as possible, an optimal influx rate per infection exists. This optimal rate is approximately three times higher than the minimal number of plasma cells providing protection. Influx rates of plasmablasts generated by vaccination approximately match this optimum level. Furthermore, the observed stability of serum concentrations of vaccine-specific antibodies implies that the influxing plasmablasts mobilize a similar number of plasma cells and that competitive infectious challenges are not more frequent than once per month.


Subject(s)
Adaptation, Biological/immunology , Immunologic Memory/immunology , Plasma Cells/immunology , Animals , Antibody Formation , Humans , Infections/immunology , Mice , Models, Immunological
10.
Breast J ; 11(1): 34-40, 2005.
Article in English | MEDLINE | ID: mdl-15647076

ABSTRACT

Individualization and optimization of risk-adapted therapy in breast cancer requires molecular profiling of individual cancer specimens. In tissues of heterogeneous cellular composition, such as mammary tissue, cancer prognostic markers and potential therapeutic target molecules are preferentially detected by in situ techniques such as immunohistochemistry. The more therapy decisions are based on immunohistochemical findings, the more histopathologists are confronted with the demand to establish standardized procedures that enable reproducible evaluation independent from the investigating laboratory. Disappointing results of recent national and international immunohistochemistry trials for steroid receptor and HER-2 analysis in breast cancer underline the need to act now beyond recommendations and well-formulated guidelines for standardization. In order to ensure staining quality in every individual immunohistochemical analysis, we have developed an on-slide control with standardized samples which are placed on every slide of a therapy-decisive stain together with the tissue of interest. Novel mini tissue microarrays were constructed comprising cell lines as standardized controls, indicating the sensitivity and reliability of the staining procedure. Standardized control material on the same slide as the patient's specimen require neither additional reagents nor time and are automatically archived on the diagnostic slide to prove the quality of staining even after years.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Histocytological Preparation Techniques/standards , Immunohistochemistry/standards , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Germany , Histocytological Preparation Techniques/methods , Humans , Immunohistochemistry/methods , Quality Control , Reference Values , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...