22 results on '"Sandra Beer-Hammer"'
Search Results
2. Human monocytic myeloid‐derived suppressor cells impair B‐cell phenotype and function in vitro
- Author
-
Annkathrin C. Teschner, Sandra Beer-Hammer, Katja Fromm, Nikolaus Rieber, Dominik Hartl, Felipe J.N. Lelis, and Jennifer Jaufmann
- Subjects
CD86 ,B-Lymphocytes ,Myeloid-Derived Suppressor Cells ,Immunology ,Biology ,Lymphocyte Activation ,Phenotype ,In vitro ,ddc ,Cell biology ,Immune system ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,Myeloid-derived Suppressor Cell ,Humans ,Immunology and Allergy ,Cells, Cultured ,B cell ,CD80 ,Cell Proliferation - Abstract
Myeloid-derived suppressor cells (MDSCs) are key regulators of immunity that initially have been defined by their ability to potently suppress T-cell responses. Recent studies collectively demonstrate that the suppressive activity of MDSCs is not limited to T cells, but rather affects a broad range of immune cell subsets. However, relatively few studies have assessed the impact of MDSCs on B cells, particularly in the human context. Here, we report that human monocytic MDSCs (M-MDSCs) significantly interfere with human B-cell proliferation and function in vitro. We further show that the inhibition occurs independent of direct cell-contact and involves the expression of suppressive mediators such as indoleamine 2, 3-dioxygenase (IDO), arginase-1 (Arg1), and nitric oxide (NO). In addition, our studies demonstrate that the suppression of B cells by M-MDSCs is paralleled by a skewing in B-cell phenotype and gene expression signatures. M-MDSCs induced the downregulation of key surface markers on activated B cells, including IgM, HLA-DR, CD80, CD86, TACI, and CD95. Concurrently, M-MDSCs but not conventional monocytes elicited alterations in the transcription of genes involved in apoptosis induction, class-switch regulation, and B-cell differentiation and function. In summary, this study expands our understanding of the regulatory role of M-MDSCs for human B-cell responses.
- Published
- 2019
- Full Text
- View/download PDF
3. The gut microbiota metabolite urolithin A inhibits NF-κB activation in LPS stimulated BMDMs
- Author
-
Khalid N M, Abdelazeem, M Zaher, Kalo, Sandra, Beer-Hammer, and Florian, Lang
- Subjects
Inflammation ,Lipopolysaccharides ,Macrophages ,Immunology ,NF-kappa B ,Pathogenesis ,Article ,Gastrointestinal Microbiome ,Toll-Like Receptor 4 ,Mice ,MicroRNAs ,RAW 264.7 Cells ,Coumarins ,Animals ,Calcium ,DNA Breaks, Double-Stranded ,Inflammation Mediators ,Reactive Oxygen Species ,Signal Transduction - Abstract
Inflammation is a natural defense process of the innate immune system, associated with the release of proinflammatory cytokines such as interleukin-1β, interleukin-6, interleukin-12 and TNFα; and enzymes including iNOS through the activation and nuclear translocation of NF-κB p65 due to the phosphorylation of IκBα. Regulation of intracellular Ca2+ is considered a promising strategy for the prevention of reactive oxygen species (ROS) production and accumulation of DNA double strand breaks (DSBs) that occurs in inflammatory-associated-diseases. Among the metabolites of ellagitannins that are produced in the gut microbiome, urolithin A (UA) has received an increasing attention as a novel candidate with anti-inflammatory and anti-oxidant effects. Here, we investigated the effect of UA on the suppression of pro-inflammatory molecules and NF-κB activation by targeting TLR4 signalling pathway. We also identified the influence of UA on Ca2+ entry, ROS production and DSBs availability in murine bone-marrow-derived macrophages challenged with lipopolysaccharides (LPS). We found that UA inhibits IκBα phosphorylation and supresses MAPK and PI3K activation. In addition, UA was able to reduce calcium entry, ROS production and DSBs availability. In conclusion, we suggest that urolithin A is a promising therapeutic agent for treating inflammatory diseases through suppression of NF-κB and preserving DNA through maintaining intracellular calcium and ROS homeostasis.
- Published
- 2020
4. Myeloid-derived suppressor cells modulate B-cell responses
- Author
-
Sandra Beer-Hammer, Iris Schäfer, Anurag Singh, Felipe J.N. Lelis, Jennifer Jaufmann, Annkathrin C. Teschner, Simone Pöschel, Katja Fromm, Nikolaus Rieber, and Dominik Hartl
- Subjects
0301 basic medicine ,Neutrophils ,medicine.medical_treatment ,Immunology ,Cell Communication ,Biology ,Lymphocyte Activation ,Nitric Oxide ,law.invention ,Nitric oxide ,Immunomodulation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,law ,medicine ,Humans ,Immunology and Allergy ,DAPI ,Cells, Cultured ,B cell ,B-Lymphocytes ,Arginase ,Myeloid-Derived Suppressor Cells ,Immunotherapy ,Acquired immune system ,Natural killer T cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Immunoglobulin M ,chemistry ,Antibody Formation ,Myeloid-derived Suppressor Cell ,Suppressor ,Reactive Oxygen Species ,Biomarkers ,030215 immunology - Abstract
Myeloid-derived suppressor cells (MDSCs) are key regulators of adaptive immunity by suppressing T-cell functions. However, their potential action on or interaction with B cells remained poorly understood. Here we demonstrate that human polymorphonuclear MDSCs differentially modulate B-cell function by suppressing B-cell proliferation and antibody production. We further demonstrate that this MDSC-mediated effect is cell contact dependent and involves established mediators such as arginase-1, nitric oxide (NO), reactive oxygen species (ROS) as well as B-cell death. Collectively, our studies provide novel evidence that human MDSCs modulate B cells, which could have future implications for immunotherapy approaches.
- Published
- 2017
- Full Text
- View/download PDF
5. CXCR1 Regulates Pulmonary Anti-Pseudomonas Host Defense
- Author
-
Barbara Moepps, Andreas Hector, Sandra Schwarz, Hasan Halit Öz, Sandra Beer-Hammer, Julie Laval, M Carevic, Carolin Schroth, Nina Frey, Manfred Kneilling, Kerstin Fuchs, Tatjana Bilich, Annika Schmidt, Kirsten Bucher, P M Murphy, Dominik Hartl, Stella E. Autenrieth, Maria Haug, Charaf Benarafa, Amit Gaggar, and J L Gao
- Subjects
0301 basic medicine ,COPD ,Lung ,Effector ,Pseudomonas aeruginosa ,Biology ,medicine.disease_cause ,medicine.disease ,Cystic fibrosis ,Microbiology ,03 medical and health sciences ,Chemokine receptor ,030104 developmental biology ,medicine.anatomical_structure ,TLR5 ,Immunity ,Immunology ,medicine ,Immunology and Allergy - Abstract
Pseudomonas aeruginosa is a key opportunistic pathogen causing disease in cystic fibrosis (CF) and other lung diseases such as chronic obstructive pulmonary disease (COPD). However, the pulmonary host defense mechanisms regulating anti-P. aeruginosa immunity remain incompletely understood. Here we demonstrate, by studying an airway P. aeruginosa infection model, in vivo bioluminescence imaging, neutrophil effector responses and human airway samples, that the chemokine receptor CXCR1 regulates pulmonary host defense against P. aeruginosa. Mechanistically, CXCR1 regulates anti-Pseudomonas neutrophil responses through modulation of reactive oxygen species and interference with Toll-like receptor 5 expression. These studies define CXCR1 as a novel, noncanonical chemokine receptor that regulates pulmonary anti-Pseudomonas host defense with broad implications for CF, COPD and other infectious lung diseases.
- Published
- 2016
- Full Text
- View/download PDF
6. Correction: mRNA-Mediated Gene Supplementation of Toll-Like Receptors as Treatment Strategy for Asthma In Vivo
- Author
-
Clara Will, Jennifer Rottenberger, Michael S. D. Kormann, Franziska Zeyer, Bernd Nürnberg, Benedikt Mothes, Dominik Hartl, Sandra Beer-Hammer, Rupert Handgretinger, and Melanie Carevic
- Subjects
Pulmonology ,Neutrophils ,lcsh:Medicine ,Pathology and Laboratory Medicine ,Immune Receptors ,Biochemistry ,White Blood Cells ,Mice ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Medicine ,Receptor ,lcsh:Science ,Immune Response ,Mice, Inbred BALB C ,Immune System Proteins ,Multidisciplinary ,biology ,Pyroglyphidae ,Toll-Like Receptors ,Animal Models ,3. Good health ,030220 oncology & carcinogenesis ,Toll ,Treatment strategy ,Female ,Cellular Types ,Research Article ,Signal Transduction ,Immune Cells ,Immunology ,Mouse Models ,Research and Analysis Methods ,03 medical and health sciences ,Signs and Symptoms ,Model Organisms ,Text mining ,Diagnostic Medicine ,In vivo ,Animals ,RNA, Messenger ,Molecular Biology Techniques ,Molecular Biology ,Gene ,Asthma ,Inflammation ,Messenger RNA ,Blood Cells ,business.industry ,lcsh:R ,Biology and Life Sciences ,Proteins ,Correction ,Cell Biology ,medicine.disease ,respiratory tract diseases ,Eosinophils ,Disease Models, Animal ,biology.protein ,lcsh:Q ,business ,030217 neurology & neurosurgery ,Cloning - Abstract
Asthma is the most common chronic disease in childhood. Although several therapeutic options are currently available to control the symptoms, many drugs have significant side effects and asthma remains an incurable disease. Microbial exposure in early life reduces the risk of asthma and several studies have suggested protective effects of Toll-like receptor (TLR) activation. We showed previously that modified mRNA provides a safe and efficient therapeutic tool for in vivo gene supplementation. Since current asthma drugs do not take patient specific immune and TLR backgrounds into consideration, treatment with tailored mRNA could be an attractive approach to account for the patient’s individual asthma phenotype. Therefore, we investigated the effect of a preventative treatment with combinations of Tlr1, Tlr2 and Tlr6 mRNA in a House Dust Mite-induced mouse model of asthma. We used chemically modified mRNA which is–in contrast to conventional viral vectors–non-integrating and highly efficient in gene transfer. In our study, we found that treatment with either Tlr1/2 mRNA or Tlr2/6 mRNA, but not Tlr2 mRNA alone, resulted in better lung function as well as reduced airway inflammation in vivo. The present results point to a potentially protective effect of TLR heterodimers in asthma pathogenesis.
- Published
- 2018
7. SLy1 regulates T-cell proliferation duringListeria monocytogenesinfection in a Foxo1-dependent manner
- Author
-
Sandra Beer-Hammer, Bernhard Reis, Daniel Schäll, Simone Brandt, and Fee Schmitt
- Subjects
Immunoprecipitation ,T cell ,Immunology ,T-cell receptor ,Signal transducing adaptor protein ,FOXO1 ,Cell cycle ,Biology ,Molecular biology ,Cell biology ,medicine.anatomical_structure ,Cytoplasm ,medicine ,Immunology and Allergy ,Phosphorylation - Abstract
Infection of mice with Listeria monocytogenes results in a strong T-cell response that is critical for an efficient defense. Here, we demonstrate that the adapter protein SLy1 (SH3-domain protein expressed in Lymphocytes 1) is essential for the generation of a fully functional T-cell response. The lack of SLy1 leads to reduced survival rates of infected mice. The increased susceptibility of SLy1 knock-out (KO) mice was caused by reduced proliferation of differentiated T cells. Ex vivo analyses of isolated SLy1 KO T cells displayed a dysregulation of Forkhead box protein O1 shuttling after TCR signaling, which resulted in an increased expression of cell cycle inhibiting genes, and therefore, reduced expansion of the T-cell population. Forkhead box protein O1 shuttles to the cytoplasm after phosphorylation in a protein complex including 14-3-3 proteins. Interestingly, we observed a similar regulation for the adapter protein SLy1, where TCR stimulation results in SLy1 phosphorylation and SLy1 export to the cytoplasm. Moreover, immunoprecipitation analyses revealed a binding of SLy1 to 14-3-3 proteins. Altogether, this study describes SLy1 as an immunoregulatory protein, which is involved in the generation of adaptive immune responses during L. monocytogenes infection, and provides a model of how SLy1 regulates T-cell proliferation.
- Published
- 2015
- Full Text
- View/download PDF
8. Innate immune system favors emergency monopoiesis at the expense of DC-differentiation to control systemic bacterial infection in mice
- Author
-
Manina Günter, Matthias Grauer, Ulrike Schleicher, Ingo B. Autenrieth, Hans-Jörg Bühring, Sandra Beer-Hammer, Lars Zender, Claudia Lengerke, Hans-Georg Rammensee, Kristin Bieber, Tilo Biedermann, Stella E. Autenrieth, Karina A. Pasquevich, and Oliver Pötz
- Subjects
Adoptive cell transfer ,Innate immune system ,medicine.medical_treatment ,Monocyte ,Immunology ,Immunosuppression ,Biology ,medicine.disease ,Sepsis ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,TLR4 ,Immunology and Allergy ,Bone marrow - Abstract
DCs are professional APCs playing a crucial role in the initiation of T-cell responses to combat infection. However, systemic bacterial infection with various pathogens leads to DC-depletion in humans and mice. The mechanisms of pathogen-induced DC-depletion remain poorly understood. Previously, we showed that mice infected with Yersinia enterocolitica (Ye) had impaired de novo DC-development, one reason for DC-depletion. Here, we extend these studies to gain insight into the molecular mechanisms of DC-depletion and the impact of different bacteria on DC-development. We show that the number of bone marrow (BM) hematopoietic progenitors committed to the DC lineage is reduced following systemic infection with different Gram-positive and Gram-negative bacteria. This is associated with a TLR4- and IFN-gamma-signaling dependent increase of committed monocyte progenitors in the BM and mature monocytes in the spleen upon Ye-infection. Adoptive transfer experiments revealed that infection-induced monopoiesis occurs at the expense of DC-development. Our data provide evidence for a general response of hematopoietic progenitors upon systemic bacterial infections to enhance monocyte production, thereby increasing the availability of innate immune cells for pathogen control, whereas impaired DC-development leads to DC-depletion, possibly driving transient immunosuppression in bacterial sepsis.
- Published
- 2015
- Full Text
- View/download PDF
9. SLy2 controls the antibody response to pneumococcal vaccine through an IL-5Rα-dependent mechanism in B-1 cells
- Author
-
Sandra Beer-Hammer, Tilo Biedermann, Andreas Hector, Fee Schmitt, Kirsten Bucher, Dominik Hartl, Michael Zemlin, Theresa Isabell Schindler, and Daniel Schäll
- Subjects
Src homology domain ,education.field_of_study ,biology ,Lymphocyte ,Immunology ,Population ,Signal transducing adaptor protein ,Molecular biology ,Immune system ,medicine.anatomical_structure ,Pneumococcal vaccine ,Immunoglobulin M ,Immunity ,biology.protein ,medicine ,Immunology and Allergy ,education - Abstract
The adaptor protein SLy2 (Src homology domain 3 lymphocyte protein 2) is located on human chromosome 21 and was reported to be among a group of genes amplified in Down's syndrome (DS) patients. DS patients characteristically show an impaired immunity to pneumococcal infections. However, molecular mechanisms linking gene amplifications with specific DS phenotypes remain elusive. To investigate the effect of SLy2 gene amplification on the mammalian immune system, we studied SLy2 overexpressing transgenic-SLy2 (TG) mice. We found that baseline immunoglobulin M (IgM) levels as well as IgM responses following Pneumovax immunizations were reduced in TG mice. Moreover, B-1 cells, the major natural IgM-producing population in mice, were reduced in the peritoneal cavity of TG mice, while other immune cell compartments were unaltered. Mechanistically, SLy2 overexpression attenuated the expression of the IL-5 receptor α chain on B-1 cells, resulting in decreased B-1 cell numbers and decreased differentiation into Ab-secreting cells. Since B-1 cells essentially contribute to immunity against Streptococcus pneumoniae, the present study provides a novel molecular link between SLy2 expression and pneumococcal-specific IgM responses in vivo. These studies suggest that the adaptor protein SLy2 is a potential future target for immunomodulatory strategies for pneumococcal infections.
- Published
- 2014
- Full Text
- View/download PDF
10. Staphylococcus aureus Phenol-Soluble Modulin Peptides Modulate Dendritic Cell Functions and Increase In Vitro Priming of Regulatory T Cells
- Author
-
Sandra Beer-Hammer, Michael Otto, Stefan Stevanovic, Stella E. Autenrieth, Dorothee Kretschmer, Andreas Peschel, Jens Schreiner, Juliane Klenk, Ji Ming Wang, and Hans-Jörg Bühring
- Subjects
Staphylococcus aureus ,Bacterial Toxins ,Immunology ,Priming (immunology) ,chemical and pharmacologic phenomena ,Biology ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Article ,Microbiology ,Proinflammatory cytokine ,Mice ,Immune system ,Animals ,Immunology and Allergy ,Formyl peptide receptor ,Chemotaxis ,FOXP3 ,Dendritic Cells ,Dendritic cell ,Acquired immune system ,Receptors, Formyl Peptide ,Clathrin ,Endocytosis ,Toll-Like Receptor 2 ,TLR2 ,Cytokines ,Female ,Peptides - Abstract
The major human pathogen Staphylococcus aureus has very efficient strategies to subvert the human immune system. Virulence of the emerging community-associated methicillin-resistant S. aureus depends on phenol-soluble modulin (PSM) peptide toxins, which are known to attract and lyse neutrophils. However, their influences on other immune cells remain elusive. In this study, we analyzed the impact of PSMs on dendritic cells (DCs) playing an essential role in linking innate and adaptive immunity. In human neutrophils, PSMs exert their function by binding to the formyl peptide receptor (FPR) 2. We show that mouse DCs express the FPR2 homolog mFPR2 as well as its paralog mFPR1 and that PSMs are chemoattractants for DCs at noncytotoxic concentrations. PSMs reduced clathrin-mediated endocytosis and inhibited TLR2 ligand-induced secretion of the proinflammatory cytokines TNF, IL-12, and IL-6, while inducing IL-10 secretion by DCs. As a consequence, treatment with PSMs impaired the capacity of DCs to induce activation and proliferation of CD4+ T cells, characterized by reduced Th1 but increased frequency of FOXP3+ regulatory T cells. These regulatory T cells secreted high amounts of IL-10, and their suppression capacity was dependent on IL-10 and TGF-β. Interestingly, the induction of tolerogenic DCs by PSMs appeared to be independent of mFPRs, as shown by experiments with mice lacking mFPR2 (mFPR2−/−) and the cognate G protein (p110γ−/−). Thus, PSMs from highly virulent pathogens affect DC functions, thereby modulating the adaptive immune response and probably increasing the tolerance toward the pathogen.
- Published
- 2013
- Full Text
- View/download PDF
11. Modified Foxp3 mRNA protects against asthma through an IL-10–dependent mechanism
- Author
-
Lauren Mays, Rupert Handgretinger, Esther von Stebut, Michael S. D. Kormann, Nikolaus Rieber, Susanne Ammon-Treiber, Dominik Hartl, Mohammed Alkhaled, Melanie Grimm, Markus Mezger, Bernd Nürnberg, Jennifer Rottenberger, Eva Müller-Hermelink, Matthias Schwab, Benedikt Mothes, Sandra Beer-Hammer, and Marco Idzko
- Subjects
Regulatory T cell ,T cell ,Thiouridine ,Gene Expression ,Cytidine ,Biology ,Transfection ,Interleukin-23 ,Cell Line ,Mice ,Th2 Cells ,medicine ,Interleukin 23 ,Animals ,Humans ,RNA, Messenger ,Mice, Knockout ,Mice, Inbred BALB C ,Innate immune system ,Airway Resistance ,Interleukin-17 ,Pyroglyphidae ,FOXP3 ,Forkhead Transcription Factors ,Genetic Therapy ,General Medicine ,T helper cell ,Asthma ,Immunity, Innate ,Interleukin-10 ,Interleukin 10 ,medicine.anatomical_structure ,Immunology ,Airway Remodeling ,Th17 Cells ,Female ,Interleukin 17 ,Inflammation Mediators ,Research Article - Abstract
Chemically modified mRNA is capable of inducing therapeutic levels of protein expression while circumventing the threat of genomic integration often associated with viral vectors. We utilized this novel therapeutic tool to express the regulatory T cell transcription factor, FOXP3, in a time- and site-specific fashion in murine lung, in order to prevent allergic asthma in vivo. We show that modified Foxp3 mRNA rebalanced pulmonary T helper cell responses and protected from allergen-induced tissue inflammation, airway hyperresponsiveness, and goblet cell metaplasia in 2 asthma models. This protection was conferred following delivery of modified mRNA either before or after the onset of allergen challenge, demonstrating its potential as both a preventive and a therapeutic agent. Mechanistically, FOXP3 induction controlled Th2 and Th17 inflammation by regulating innate immune cell recruitment through an IL-10-dependent pathway. The protective effects of FOXP3 could be reversed by depletion of IL-10 or administration of recombinant IL-17A or IL-23. Delivery of Foxp3 mRNA to sites of inflammation may offer a novel, safe therapeutic tool for the treatment of allergic asthma and other diseases driven by an imbalance in helper T cell responses.
- Published
- 2013
- Full Text
- View/download PDF
12. mRNA-Mediated Gene Supplementation of Toll-Like Receptors as Treatment Strategy for Asthma In Vivo
- Author
-
Jennifer Rottenberger, Franziska Zeyer, Clara Will, Melanie Carevic, Sandra Beer-Hammer, Bernd Nürnberg, Benedikt Mothes, Michael S. D. Kormann, Dominik Hartl, and Rupert Handgretinger
- Subjects
0301 basic medicine ,Allergy ,Multidisciplinary ,lcsh:R ,lcsh:Medicine ,Inflammation ,Biology ,medicine.disease ,3. Good health ,respiratory tract diseases ,Pathogenesis ,03 medical and health sciences ,TLR2 ,030104 developmental biology ,Immune system ,In vivo ,TLR6 ,Immunology ,medicine ,lcsh:Q ,medicine.symptom ,lcsh:Science ,Asthma - Abstract
Asthma is the most common chronic disease in childhood. Although several therapeutic options are currently available to control the symptoms, many drugs have significant side effects and asthma remains an incurable disease. Microbial exposure in early life reduces the risk of asthma and several studies have suggested protective effects of Toll-like receptor (TLR) activation. We showed previously that modified mRNA provides a safe and efficient therapeutic tool for in vivo gene supplementation. Since current asthma drugs do not take patient specific immune and TLR backgrounds into consideration, treatment with tailored mRNA could be an attractive approach to account for the patient’s individual asthma phenotype. Therefore, we investigated the effect of a preventative treatment with combinations of Tlr1, Tlr2 and Tlr6 mRNA in a House Dust Mite-induced mouse model of asthma. We used chemically modified mRNA which is–in contrast to conventional viral vectors–non-integrating and highly efficient in gene transfer. In our study, we found that treatment with either Tlr1/2 mRNA or Tlr2/6 mRNA, but not Tlr2 mRNA alone, resulted in better lung function as well as reduced airway inflammation in vivo. The present results point to a potentially protective effect of TLR heterodimers in asthma pathogenesis.
- Published
- 2016
13. Cell-intrinsic and -extrinsic control of Treg-cell homeostasis and function revealed by inducedCD28deletion
- Author
-
Tea Gogishvili, Sandra Beer-Hammer, Klaus Pfeffer, Fred Lühder, Thomas Hünig, and Sandra Goebbels
- Subjects
0303 health sciences ,medicine.medical_treatment ,Immunology ,Cell ,CD28 ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,Cell biology ,Thymectomy ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,Immunology and Allergy ,IL-2 receptor ,Gene ,Function (biology) ,Homeostasis ,030304 developmental biology ,030215 immunology - Abstract
While the requirement for CD28 and its ligands for the generation and function of “natural” (n)Treg cells is well established, it has not been possible yet to investigate cell-intrinsic effects after interrupted CD28 expression. Here, we demonstrate a selective loss of Treg cells after disruption of the CD28 gene. The decline in Treg-cell number was accompanied by reduced homeostatic proliferation, probably due to lack of costimulation during self-antigen recognition, and by impaired Treg-cell function including downregulation of CTLA-4. The decline in Treg-cell number was unaffected by thymectomy or by the presence of CD28 expressing T cells within the same animal, indicating that impairment of peripheral homeostasis and function of nTreg cells by CD28 deletion is cell-intrinsic. In contrast, downregulation of CD25, the α chain of the IL-2R, did not occur in the presence of WT T cells, indicating that its expression does not depend on CD28 signals in cis.
- Published
- 2012
- Full Text
- View/download PDF
14. Defective Macrophage Migration in Gαi2- but Not Gαi3-Deficient Mice
- Author
-
Ana Novakovic, Reinhold E. Schmidt, Syed Raza Ali, Duc D. Le, Roland P. Piekorz, Sandra Beer-Hammer, Bernd Nürnberg, Shahzad N. Syed, Kristina Wiege, and J. Engelbert Gessner
- Subjects
Lipopolysaccharides ,Mice, 129 Strain ,medicine.medical_treatment ,Acute Lung Injury ,Immunology ,Motility ,GTP-Binding Protein alpha Subunits, Gi-Go ,Peritonitis ,Biology ,Lung injury ,Monocytes ,Mice ,Heterotrimeric G protein ,medicine ,Animals ,Immunology and Allergy ,Mice, Knockout ,Macrophages ,Chemotaxis ,Cell Migration Inhibition ,Cell migration ,Transfection ,Molecular biology ,Cell biology ,Cytokine ,Thioglycolates ,GTP-Binding Protein alpha Subunit, Gi2 - Abstract
Various heterotrimeric Gi proteins are considered to be involved in cell migration and effector function of immune cells. The underlying mechanisms, how they control the activation of myeloid effector cells, are not well understood. To elucidate isoform-redundant and -specific roles for Gαi proteins in these processes, we analyzed mice genetically deficient in Gαi2 or Gαi3. First, we show an altered distribution of tissue macrophages and blood monocytes in the absence of Gαi2 but not Gαi3. Gαi2-deficient but not wild-type or Gαi3-deficient mice exhibited reduced recruitment of macrophages in experimental models of thioglycollate-induced peritonitis and LPS-triggered lung injury. In contrast, genetic ablation of Gαi2 had no effect on Gαi-dependent peritoneal cytokine production in vitro and the phagocytosis-promoting function of the Gαi-coupled C5a anaphylatoxin receptor by liver macrophages in vivo. Interestingly, actin rearrangement and CCL2- and C5a anaphylatoxin receptor-induced chemotaxis but not macrophage CCR2 and C5a anaphylatoxin receptor expression were reduced in the specific absence of Gαi2. Furthermore, knockdown of Gαi2 caused decreased cell migration and motility of RAW 264.7 cells, which was rescued by transfection of Gαi2 but not Gαi3. These results indicate that Gαi2, albeit redundant to Gαi3 in some macrophage activation processes, clearly exhibits a Gαi isoform-specific role in the regulation of macrophage migration.
- Published
- 2012
- Full Text
- View/download PDF
15. Fluorescent Ly6G antibodies determine macrophage phagocytosis of neutrophils and alter the retrieval of neutrophils in mice
- Author
-
Sandra Beer-Hammer, Fee Schmitt, Katja Schenke-Layland, Bernd Nürnberg, Inken Dillmann, Kirsten Bucher, Stella E. Autenrieth, and Publica
- Subjects
Adoptive cell transfer ,Neutrophils ,Phagocytosis ,Immunology ,Spleen ,Bone Marrow Cells ,Biology ,Endocytosis ,Antibodies ,Fluorescence ,Flow cytometry ,In vivo ,medicine ,Immunology and Allergy ,Macrophage ,Animals ,Antigens, Ly ,Fluorescent Dyes ,Binding Sites ,medicine.diagnostic_test ,Macrophages ,Phycocyanin ,Phycoerythrin ,Cell Biology ,Flow Cytometry ,Molecular biology ,Adoptive Transfer ,Mice, Inbred C57BL ,medicine.anatomical_structure ,biology.protein ,Antibody ,Fluorescein-5-isothiocyanate - Abstract
Fluorescently labeled Ly6G antibodies enable the tracking of neutrophils in mice, whereas purified anti-Ly6G rapidly depletes neutrophils from the circulation. The mechanisms underlying neutrophil depletion are still under debate. Here, we examined how identical Ly6G antibodies coupled to different fluorochromes affect neutrophil fate in vivo. BM cells stained with Ly6G antibodies were injected into mice. The number of retrieved anti-Ly6G-FITC+ cells was reduced significantly in comparison with anti-Ly6G-APC+ or anti-Ly6G-PE+ cells. Flow cytometry and multispectral imaging flow cytometry analyses revealed that anti-Ly6G-FITC+ neutrophils were preferentially phagocytosed by BMMs in vitro and by splenic, hepatic, and BM macrophages in vivo. Direct antibody injection of anti-Ly6G-FITC but not anti-Ly6G-PE depleted neutrophils to the same degree as purified anti-Ly6G, indicating that the FITC-coupled antibody eliminates neutrophils by a similar mechanism as the uncoupled antibody. With the use of a protein G-binding assay, we demonstrated that APC and PE but not FITC coupling inhibited access to interaction sites on the anti-Ly6G antibody. We conclude the following: 1) that neutrophil phagocytosis by macrophages is a central mechanism in anti-Ly6G-induced neutrophil depletion and 2) that fluorochrome-coupling can affect functional properties of anti-Ly6G antibodies, thereby modifying macrophage uptake of Ly6G-labeled neutrophils and neutrophil retrieval following adoptive cell transfer or injection of fluorescent anti-Ly6G.
- Published
- 2014
16. Deficiency of the adaptor protein SLy1 results in a natural killer cell ribosomopathy affecting tumor clearance
- Author
-
Hosseinali Asgharian, Andrew E. Gelman, Reza Ghasemi, Alexander S. Krupnick, Hani S. Zaher, Yizhan Guo, Jinsheng Yu, Wenjun Li, Ryuji Higashikubo, Daniel Kreisel, Anthony R. French, Sandra Beer-Hammer, Daniel Schäll, Beatrice Plougastel-Douglas, Leonard B. Maggi, Stephanie Chang, Alex Zheleznyak, Wayne M. Yokoyama, and Saeed Arefanian
- Subjects
0301 basic medicine ,natural killer cells ,Lymphokine-activated killer cell ,Ribosomopathy ,Immunosurveillance ,Immunology ,Cell ,Signal transducing adaptor protein ,ribosomopathy ,Biology ,3. Good health ,Natural killer cell ,lung cancer ,03 medical and health sciences ,Interleukin 21 ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cytoplasm ,medicine ,Immunology and Allergy ,Signal transduction ,Original Research - Abstract
Individuals with robust natural killer (NK) cell function incur lower rates of malignancies. To expand our understanding of genetic factors contributing to this phenomenon, we analyzed NK cells from cancer resistant and susceptible strains of mice. We identified a correlation between NK levels of the X-chromosome-located adaptor protein SLy1 and immunologic susceptibility to cancer. Unlike the case for T or B lymphocytes, where SLy1 shuttles between the cytoplasm and nucleus to facilitate signal transduction, in NK cells SLy1 functions as a ribosomal protein and is located solely in the cytoplasm. In its absence, ribosomal instability results in p53-mediated NK cell senescence and decreased clearance of malignancies. NK defects are reversible under inflammatory conditions and viral clearance is not impacted by SLy1 deficiency. Our work defines a previously unappreciated X-linked ribosomopathy that results in a specific and subtle NK cell dysfunction leading to immunologic susceptibility to cancer.
- Published
- 2016
- Full Text
- View/download PDF
17. Interruption of CD28-mediated costimulation during allergen challenge protects mice from allergic airway disease
- Author
-
Fred Lühder, Natalie E. Nieuwenhuizen, Frank Brombacher, Klaus Pfeffer, Thomas Hünig, Sandra Beer-Hammer, Frank Kirstein, Tea Gogishvili, Christian Taube, Sebastian Reuter, and Sandra Goebbels
- Subjects
Allergic asthma ,Lymphocyte Activation ,Immunoglobulin E ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Mice ,0302 clinical medicine ,Airway resistance ,Allergen ,Immunology and Allergy ,Sensitization ,Mice, Inbred BALB C ,0303 health sciences ,biology ,Antibodies, Monoclonal ,ovalbumin ,respiratory system ,3. Good health ,medicine.anatomical_structure ,costimulation ,conditional CD28 knockout mice ,Female ,medicine.symptom ,Immunology ,Inflammation ,03 medical and health sciences ,Th2 Cells ,CD28 Antigens ,Respiratory Hypersensitivity ,medicine ,Animals ,Humans ,Antigens, Dermatophagoides ,Lymphocyte Count ,Antibodies, Blocking ,030304 developmental biology ,House dust mite ,CD28-specific mAb ,business.industry ,Receptor Cross-Talk ,Airway obstruction ,medicine.disease ,biology.organism_classification ,Mice, Mutant Strains ,respiratory tract diseases ,Disease Models, Animal ,CTLA-4 ,biology.protein ,business ,030215 immunology - Abstract
Background Allergic asthma is a T H 2-promoted hyperreactivity with an immediate, IgE, and mast cell–dependent response followed by eosinophil-dominated inflammation and airway obstruction. Objective Because costimulation by CD28 is essential for T H 2 but not T H 1 responses, we investigated the effect of selective interference with this pathway in mice using the models of ovalbumin and house dust mite–induced airway inflammation. Methods To study the role of CD28 in the effector phase of allergic airway inflammation, we developed an inducibly CD28-deleting mouse strain or alternatively used a CD28 ligand-binding site–specific mouse anti-mouse mAb blocking CD28 engagement. Results We show that even after systemic sensitization to the allergen, interruption of CD28-mediated costimulation is highly effective in preventing airway inflammation during challenge. In addition to improving airway resistance and histopathologic presentation and reducing inflammatory infiltrates, antibody treatment during allergen challenge resulted in a marked relative increase in regulatory T-cell numbers among the CD4 T-cell subset of the challenged lung. Conclusion Selective interference with CD28-mediated costimulation during allergen exposure might be an attractive therapeutic concept for allergic asthma.
- Published
- 2012
- Full Text
- View/download PDF
18. Gαi2 is the essential Gαi protein in immune complex-induced lung disease
- Author
-
Syed Raza Ali, J. Engelbert Gessner, Britta Gewecke, Roland P. Piekorz, Kristina Wiege, Jörg Reutershan, Reinhold E. Schmidt, Ana Novakovic, Sandra Beer-Hammer, Franziska M. Konrad, Bernd Nürnberg, and Katja Pexa
- Subjects
Chemokine ,Mice, 129 Strain ,Endothelium ,Neutrophils ,Immunology ,Acute Lung Injury ,Down-Regulation ,Inflammation ,GTP-Binding Protein alpha Subunits, Gi-Go ,C5a receptor ,Mice ,Heterotrimeric G protein ,medicine ,Arthus Reaction ,Cell Adhesion ,Immunology and Allergy ,Animals ,Mice, Knockout ,biology ,Chemotaxis ,Neutrophil extracellular traps ,Cell biology ,Up-Regulation ,Mice, Inbred C57BL ,Chemotaxis, Leukocyte ,Disease Models, Animal ,medicine.anatomical_structure ,biology.protein ,Endothelium, Vascular ,medicine.symptom ,Signal transduction ,GTP-Binding Protein alpha Subunit, Gi2 - Abstract
Heterotrimeric G proteins of the Gαi family have been implicated in signaling pathways regulating cell migration in immune diseases. The Gαi-protein–coupled C5a receptor is a critical regulator of IgG FcR function in experimental models of immune complex (IC)–induced inflammation. By using mice deficient for Gαi2 or Gαi3, we show that Gαi2 is necessary for neutrophil influx in skin and lung Arthus reactions and agonist-induced neutrophilia in the peritoneum, whereas Gαi3 plays a less critical but variable role. Detailed analyses of the pulmonary IC-induced inflammatory response revealed several shared functions of Gαi2 and Gαi3, including mediating C5a anaphylatoxin receptor–induced activation of macrophages, involvement in alveolar production of chemokines, transition of neutrophils from bone marrow into blood, and modulation of CD11b and CD62L expression that account for neutrophil adhesion to endothelial cells. Interestingly, C5a-stimulated endothelial polymorphonuclear neutrophil transmigration, but not chemotaxis, is enhanced versus reduced in the absence of neutrophil Gαi3 or Gαi2, respectively, and knockdown of endothelial Gαi2 caused decreased transmigration of wild-type neutrophils. These data demonstrate that Gαi2 and Gαi3 contribute to inflammation by redundant, overlapping, and Gαi-isoform–specific mechanisms, with Gαi2 exhibiting unique functions in both neutrophils and endothelial cells that appear essential for polymorphonuclear neutrophil recruitment in IC disease.
- Published
- 2012
19. Depletion of dendritic cells enhances innate anti-bacterial host defense through modulation of phagocyte homeostasis
- Author
-
Karina A. Pasquevich, Sandra Beer-Hammer, Guido H. Wabnitz, Stella E. Autenrieth, Kristin Hochweller, Manina Günter, Natalio Garbi, Doreen Drechsler, Günter J. Hämmerling, Philipp Warnke, Ana Novakovic, Cecilia S.M. Lucero Estrada, Ingo B. Autenrieth, and Yvonne Samstag
- Subjects
Adoptive cell transfer ,Phagocyte ,Neutrophils ,Cell Separation ,purl.org/becyt/ford/1 [https] ,Mice ,Homeostasis ,lcsh:QH301-705.5 ,Cells, Cultured ,Phagocytes ,Acquired immune system ,Adoptive Transfer ,Up-Regulation ,Bacterial Pathogens ,medicine.anatomical_structure ,Infectious Diseases ,Medicine ,Female ,CIENCIAS NATURALES Y EXACTAS ,Research Article ,lcsh:Immunologic diseases. Allergy ,PHAGOCITES ,Yersinia Infections ,Immune Cells ,Immunology ,Spleen ,Mice, Transgenic ,Biology ,DENDRITIC CELLS ,Microbiology ,Ciencias Biológicas ,Immune system ,Biología Celular, Microbiología ,Immunity ,Virology ,Genetics ,medicine ,Splenocyte ,Animals ,purl.org/becyt/ford/1.6 [https] ,Molecular Biology ,Yersinia enterocolitica ,Innate immune system ,Bacteria ,Dendritic Cells ,Immunity, Innate ,lcsh:Biology (General) ,Immune System ,Parasitology ,lcsh:RC581-607 - Abstract
Dendritic cells (DCs) as professional antigen-presenting cells play an important role in the initiation and modulation of the adaptive immune response. However, their role in the innate immune response against bacterial infections is not completely defined. Here we have analyzed the role of DCs and their impact on the innate anti-bacterial host defense in an experimental infection model of Yersinia enterocolitica (Ye). We used CD11c-diphtheria toxin (DT) mice to deplete DCs prior to severe infection with Ye. DC depletion significantly increased animal survival after Ye infection. The bacterial load in the spleen of DC-depleted mice was significantly lower than that of control mice throughout the infection. DC depletion was accompanied by an increase in the serum levels of CXCL1, G-CSF, IL-1α, and CCL2 and an increase in the numbers of splenic phagocytes. Functionally, splenocytes from DC-depleted mice exhibited an increased bacterial killing capacity compared to splenocytes from control mice. Cellular studies further showed that this was due to an increased production of reactive oxygen species (ROS) by neutrophils. Adoptive transfer of neutrophils from DC-depleted mice into control mice prior to Ye infection reduced the bacterial load to the level of Ye-infected DC-depleted mice, suggesting that the increased number of phagocytes with additional ROS production account for the decreased bacterial load. Furthermore, after incubation with serum from DC-depleted mice splenocytes from control mice increased their bacterial killing capacity, most likely due to enhanced ROS production by neutrophils, indicating that serum factors from DC-depleted mice account for this effect. In summary, we could show that DC depletion triggers phagocyte accumulation in the spleen and enhances their anti-bacterial killing capacity upon bacterial infection., Author Summary Dendritic cells (DCs) are professional antigen-presenting cells playing a crucial role in the initiation of T-cell responses to combat infection. DCs adapt their immune response according to the type of pathogen. For example, in response to intracellular bacteria, DCs produce IL-12, thereby initiating Th1 polarization, whereas in response to extracellular parasites or extracellular bacteria, DCs instruct Th2 or Th17 polarization, respectively. Nevertheless, their role in innate immunity is less well understood. To address this, we studied the role of DCs upon infection with the Gram-negative enteropathogenic bacteria Yersinia enterocolitica (Ye) and used a mouse model to deplete DCs. We found that DCs have an unexpected role during severe infection as depletion of these cells resulted in better outcome of infection as well as less bacterial load. We also found that DC depletion increased the number of phagocytes with improved anti-bacterial capacity in the spleen. Our study provides new insights into the role of DCs in innate immune response upon bacterial infection and points towards a complex interaction between DCs and phagocyte homeostasis. DC alteration during infection might also be an interesting target for immunotherapy in the future to guide the outcome of infection.
- Published
- 2011
20. The catalytic PI3K isoforms p110{gamma} and p110{delta} contribute to B cell development and maintenance, transformation, and proliferation
- Author
-
Philipp Dresing, Klaus Pfeffer, Bernhard Reis, Bernd Nürnberg, Emilio Hirsch, Roland P. Piekorz, Veronika Sexl, Judith Alferink, Daniel Degrandi, Eva Zebedin, Sandra Beer-Hammer, Stefanie Scheu, and Max von Holleben
- Subjects
Male ,Adoptive cell transfer ,lymphoid cells ,Class I Phosphatidylinositol 3-Kinases ,Cellular differentiation ,Immunology ,Naive B cell ,Abelson murine leukemia virus ,Blotting, Western ,redundancy ,Abelson oncogene ,survival ,differentiation ,phosphoinositide 3-kinase ,Biology ,Genes, abl ,CD19 ,Mice ,Phosphatidylinositol 3-Kinases ,Bone Marrow ,medicine ,Immunology and Allergy ,Animals ,Class Ib Phosphatidylinositol 3-Kinase ,RNA, Messenger ,PI3K/AKT/mTOR pathway ,B cell ,Cell Proliferation ,Mice, Knockout ,B-Lymphocytes ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Differentiation ,Cell Biology ,Flow Cytometry ,Adoptive Transfer ,Cell biology ,Isoenzymes ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,P110δ ,biology.protein ,Female ,Signal Transduction - Abstract
PI3K isoforms p110γ and p110δ are required for early B cell development, peripheral maintenance, transformation by the Abelson oncogene, and LPS-induced proliferation. Class I PI3K-dependent signaling regulates cell proliferation, differentiation, and survival. Analysis of gene-deficient mice revealed specific roles for the hematopoietically expressed PI3K catalytic subunits, p110γ and p110δ, in development and function of T and B lymphocytes. However, the functional redundancy between these two PI3K isoforms in the B cell lineage remains unclear. Here, we demonstrate that p110δ and p110γ are expressed in B cells at early developmental stages. Normal B cell differentiation requires both isoforms, as p110γ/p110δ double deficiency causes an increased percentage of CD43hi/B220+/CD19− cells as compared with single deficiency. Interestingly, initial transformation efficiency of B cell precursors was strongly reduced in double-deficient cells following transformation by p185 bcr-abl or v-abl oncogenes as compared with single-deficient cells. The requirement of p110γ and p110δ in B cell development is underlined by reduced splenic B cell numbers of p110γ/p110δ double-deficient mice and of lethally irradiated wild-type mice reconstituted with double-deficient BM. Moreover, the peripheral maintenance of p110γ/p110δ double-deficient T and B cells was highly impaired following adoptive transfer of double-deficient splenocytes into wild-type mice. Functionally, LPS stimulation of splenocytes revealed proliferation defects resulting in decreased survival of p110γ/p110δ double-deficient B cells, which correlated with impaired induction of D-type cyclins and Bcl-XL. Surprisingly, this was not observed when purified B cells were analyzed, indicating a contribution of likely cell-extrinsic factor(s) to the impaired proliferation of double-deficient B cells. Thus, we provide novel evidence that p110γ and p110δ have overlapping and cell-extrinsic roles in the development, peripheral maintenance, and function of B cells.
- Published
- 2010
21. The orphan adapter protein SLY1 as a novel anti-apoptotic protein required for thymocyte development
- Author
-
Bernhard Reis, Sandra Beer-Hammer, and Klaus Pfeffer
- Subjects
lcsh:Immunologic diseases. Allergy ,Cellular differentiation ,Lymphocyte ,CD8 Antigens ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,Apoptosis ,Thymus Gland ,Lymphocyte Activation ,Cell Line ,Inhibitor of Apoptosis Proteins ,Mice ,medicine ,Animals ,B cell ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Precursor Cells, T-Lymphoid ,Membrane Glycoproteins ,biology ,Receptors, Notch ,GRB10 ,Signal transducing adaptor protein ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Molecular biology ,Cell biology ,Mice, Inbred C57BL ,Thymocyte ,Adaptor Proteins, Vesicular Transport ,medicine.anatomical_structure ,Cell culture ,CD4 Antigens ,biology.protein ,Signal transduction ,lcsh:RC581-607 ,Research Article ,Signal Transduction - Abstract
BackgroundSH3 containing Lymphocyte Protein (SLY1) is a putative adapter protein exclusively expressed in lymphocytes which is involved in antigen receptor induced activation. We previously have generated SLY1Δ/Δmice harbouring a partial deletion in the N-terminal region of SLY1 which revealed profound immunological defects in T and B cell functions.ResultsIn this study, T cell development in SLY1-/-and SLY1Δ/Δmice was analysedex vivoand upon cultivation with the bone marrow stromal cell line OP9. SLY1-deficient thymocytes were compromised in inducing nutrient receptor expression and ribosomal protein S6 phosphorylation, indicating a defect in mTOR complex activation. Furthermore, SLY1 was identified as a novel anti-apoptotic protein required for developmental progression of T cell precursors to the CD4+CD8+double-positive stage by protecting from premature programmed cell death initiation in developing CD4-CD8-double-negative thymocytes. In addition, SLY1 phosphorylation was differentially regulated upon Notch ligand-mediated stimulation and expression of the preTCR.ConclusionThus, our results suggest a non-redundant role for SLY1 in integrating signals from both receptors in early T cell progenitors in the thymus.
- Published
- 2009
22. The Innate Immune System Favors Emergency Monopoiesis at the Expense of DC Differentiation to Control Bacterial Infections
- Author
-
Sandra Beer-Hammer, Lars Zender, Manina Günter, Stella E. Autenrieth, Ingo B. Autenrieth, Tilo Biedermann, Karina A. Pasquevich, Claudia Lengerke, Hans-Georg Rammensee, Hans-Jörg Bühring, Kristin Bieber, Oliver Pötz, Lothar Kanz, Matthias Grauer, and Ulrike Schleicher
- Subjects
Chemokine ,Innate immune system ,Myeloid ,Monocyte ,Immunology ,Spleen ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Acquired immune system ,Biochemistry ,medicine.anatomical_structure ,Immune system ,Monocytosis ,medicine ,biology.protein - Abstract
Dendritic cells (DCs) are critical in host defense against infection, bridging the innate and adaptive immune system. Patients with sepsis display reduced circulating and splenic DCs and impaired DC function that may contribute to prolonged immune suppression and exacerbation of infection. However, the mechanisms of pathogen-induced DC depletion remain poorly understood. Here, a mouse model of systemic bacterial infection was employed to analyze the impact of different bacterial pathogens on DC development in vivo. We found that the numbers of bone marrow (BM) hematopoietic progenitors committed to the DC lineages were reduced following systemic infection with different Gram-positive and Gram-negative bacteria. In parallel, a TLR4-dependent increase of committed monocyte progenitors in the BM as well as mature monocytes in the spleen was observed. In line, adoptively transferred FLT3+ myeloid progenitors (MPs) developed preferentially to monocytes at the expense of DCs in infected animals. Analyses performed on mixed BM chimeras suggested that both the reduction of DC progenitors and the induction of monopoiesis following infection were dependent on extrinsic TLR4 signaling driving the secretion of IFN-g regulated chemokines. Consistently, these effects were completely abrogated by suppression of IFN-g signaling. Elevated monocyte numbers in the spleen triggered by infection were due to a CCR2-dependent egress from the BM. In CCR2-deficient mice, in which monocytosis reportedly is abrogated, we observed a significantly increased bacterial load in the spleen and a reduced survival rate, highlighting the importance of monocytes for bacterial clearance. Together, our data provide evidence for a general response of myeloid progenitors upon bacterial infection to enhance monocyte production, thereby increasing the availability of innate immune cells as a first line of defense against invading pathogens. Concomitantly the development of DCs is impaired, which may be responsible for transient immunosuppression in e.g. bacterial sepsis. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.