Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Manfred B. Lutz is active.

Publication


Featured researches published by Manfred B. Lutz.


Journal of Immunological Methods | 1999

An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow.

Manfred B. Lutz; Nicole A. Kukutsch; A. L. J. Ogilvie; Susanne Rössner; Franz Koch; Nikolaus Romani; Gerold Schuler

As dendritic cells (DC) are rare populations in all organs, their generation from hematopoietic precursors in large quantities has proven critical to study their biology. From murine bone marrow about 5 x 10(6) cells at 70% purity are obtained per mouse after 8 days of culture with GM-CSF. We have improved this standard method and routinely achieve a 50-fold higher yield, i.e., 1-3 x 10(8) immature and mature DC per mouse at 90-95% purity. The major modifications were: (i) the avoidance of any active depletion of bone marrow cell subpopulations to circumvent loss of precursors, (ii) a lower plating density of bone marrow cells, (iii) a prolonged culture period of 10-12 days, (iv) the reduction of the GM-CSF dose from day 8 or 10 onwards to reduce granulocyte contaminations. The final non-adherent population at day 10-12 constitutes a mixture of immature and mature DC. Further maturation of DC could be induced by high doses of LPS or TNF-alpha for the last 24 h, where 50-70% of the non-adherent fraction represented mature DC with high levels of NLDC-145, CD86 and CD40. This method allows by simple means the generation of high numbers of murine DC with very low B cell or granulocyte contaminations. It will be valuable to study DC biology notably at the molecular level.


Trends in Immunology | 2002

Immature, semi-mature and fully mature dendritic cells: which signals induce tolerance or immunity?

Manfred B. Lutz; Gerold Schuler

Dendritic cells (DCs) are currently divided into tolerogenic immature and immunogenic mature differentiation stages. However, recent findings challenge this model by reporting mature DCs as inducers of regulatory CD4+ T cells in vivo. This implies that decisive tolerogenic and immunogenic maturation signals for DCs might exist. Closer inspection reveals that tolerance is observed when partial- or semi-maturation of DCs occurs, whereas only full DC maturation is immunogenic. The decisive immunogenic signal seems to be the release of proinflammatory cytokines from the DCs. Moreover, the semi-mature DC phenotype is comparable to steady-state migratory veiled DCs within the lymphatics, which seem to continuously tolerize lymph node T cells against tissue-derived self-antigens or apoptotic cells.


Blood | 2010

Nomenclature of monocytes and dendritic cells in blood

Loems Ziegler-Heitbrock; Petronela Ancuta; Suzanne M. Crowe; Marc Dalod; Veronika Grau; Derek N. J. Hart; Pieter J. M. Leenen; Yong-Jun Liu; G. Gordon MacPherson; Gwendalyn J. Randolph; Juergen E. Scherberich; Juergen Schmitz; Ken Shortman; Silvano Sozzani; Herbert Strobl; Marek Zembala; Jonathan M. Austyn; Manfred B. Lutz

Monocytes and cells of the dendritic cell lineage circulate in blood and eventually migrate into tissue where they further mature and serve various functions, most notably in immune defense. Over recent years these cells have been characterized in detail with the use of cell surface markers and flow cytometry, and subpopulations have been described. The present document proposes a nomenclature for these cells and defines 3 types of monocytes (classical, intermediate, and nonclassical monocytes) and 3 types of dendritic cells (plasmacytoid and 2 types of myeloid dendritic cells) in human and in mouse blood. This classification has been approved by the Nomenclature Committee of the International Union of Immunological Societies, and we are convinced that it will facilitate communication among experts and in the wider scientific community.


Journal of Experimental Medicine | 2002

Repetitive Injections of Dendritic Cells Matured with Tumor Necrosis Factor α Induce Antigen-specific Protection of Mice from Autoimmunity

Mauritius Menges; Susanne Rößner; Constanze Voigtländer; Heike Schindler; Nicole A. Kukutsch; Christian Bogdan; Klaus J. Erb; Gerold Schuler; Manfred B. Lutz

Mature dendritic cells (DCs) are believed to induce T cell immunity, whereas immature DCs induce T cell tolerance. Here we describe that injections of DCs matured with tumor necrosis factor (TNF)-α (TNF/DCs) induce antigen-specific protection from experimental autoimmune encephalomyelitis (EAE) in mice. Maturation by TNF-α induced high levels of major histocompatibility complex class II and costimulatory molecules on DCs, but they remained weak producers of proinflammatory cytokines. One injection of such TNF/DCs pulsed with auto-antigenic peptide ameliorated the disease score of EAE. This could not be observed with immature DCs or DCs matured with lipopolysaccharide (LPS) plus anti-CD40. Three consecutive injections of peptide-pulsed TNF/DCs derived from wild-type led to the induction of peptide-specific predominantly interleukin (IL)-10–producing CD4+ T cells and complete protection from EAE. Blocking of IL-10 in vivo could only partially restore the susceptibility to EAE, suggesting an important but not exclusive role of IL-10 for EAE prevention. Notably, the protection was peptide specific, as TNF/DCs pulsed with unrelated peptide could not prevent EAE. In conclusion, this study describes that stimulation by TNF-α results in incompletely matured DCs (semi-mature DCs) which induce peptide-specific IL-10–producing T cells in vivo and prevent EAE.


Journal of Experimental Medicine | 2002

Human CD4+CD25+ Regulatory, Contact-dependent T Cells Induce Interleukin 10–producing, Contact-independent Type 1-like Regulatory T Cells

Detlef Dieckmann; Cord Henrik Bruett; Heidi Ploettner; Manfred B. Lutz; Gerold Schuler

It has been recently demonstrated that regulatory CD4+CD25+ CD45RO+ T cells are present in the peripheral blood of healthy adults and exert regulatory function similar to their rodent counterparts. It remains difficult to understand how the small fraction of these T cells that regulate via direct cell-to-cell contact and not via secretion of immunosuppressive cytokines could mediate strong immune suppression. Here we show that human CD4+CD25+ T cells induce long-lasting anergy and production of interleukin (IL)-10 in CD4+CD25− T cells. These anergized CD4+CD25− T cells then suppress proliferation of syngenic CD4+ T cells via IL-10 but independent of direct cell contact, similar to the so-called type 1 regulatory T (Tr1) cells. This ‘catalytic’ function of CD4+CD25+ T cells to induce Tr1-like cells helps to explain their central role for the maintenance of immune homeostasis.


European Journal of Immunology | 2000

IMMATURE DENDRITIC CELLS GENERATED WITH LOW DOSES OF GM-CSF IN THE ABSENCE OF IL-4 ARE MATURATION RESISTANT AND PROLONG ALLOGRAFT SURVIVAL IN VIVO

Manfred B. Lutz; R M Suri; M Niimi; A. L. J. Ogilvie; Nicole A. Kukutsch; Susanne Rössner; Gerold Schuler; Jonathan M. Austyn

Dendritic cells (DC) were cultured from mouse bone marrow (BM) progenitors in low concentrations of granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) (GMlo DC) by two different protocols. The phenotype and functional properties of these GMlo DC were compared to those of standard BM‐DC cultures generated in high concentrations of GM‐CSF (GMhi DC) or in low GM‐CSF plus IL‐4 (GMlo/IL‐4 DC). An effect of IL‐4 on maturation was observed only at low but not high doses of GM‐CSF. Compared to mature DC, GMlo DC were phenotypically immature, weak stimulators of allogeneic and peptide‐specific T cell responses, but substantially more potent in presentation of native protein. Immature GMlo DC were resistant to maturation by lipopolysaccharide, TNF‐α or anti‐CD40 monoclonal antibodies, as the expression of co‐stimulatory molecules was not increased, and stimulatory activity in oxidative mitogenesis was not enhanced. These maturation‐resistant immature GMlo DC induced T cell unresponsiveness in vitro and in vivo. GMlo DC also prolonged haplotype‐specific cardiac allograft survival (from 8 days to >100 days median survival time) when they were administered 7 days (but not 3, 14 or 28 days) before transplantation. Our findings may have important implications for future studies in T cell tolerance induction in vivo.


Journal of Immunology | 2002

Matrix Metalloproteinases 9 and 2 Are Necessary for the Migration of Langerhans Cells and Dermal Dendritic Cells from Human and Murine Skin

Gudrun Ratzinger; Patrizia Stoitzner; Susanne Ebner; Manfred B. Lutz; Guy T. Layton; Christian Rainer; Robert M. Senior; J. Michael Shipley; Peter Fritsch; Gerold Schuler; Nikolaus Romani

Dendritic cells migrate from the skin to the draining lymph nodes. They transport immunogenic MHC-peptide complexes, present them to Ag-specific T cells in the T areas, and thus generate immunity. Migrating dendritic cells encounter physical obstacles, such as basement membranes and collagen meshwork. Prior work has revealed that matrix metalloproteinase-9 (MMP-9) contributes to mouse Langerhans cell migration. In this study, we use mouse and human skin explant culture models to further study the role of MMPs in the migration and maturation of skin dendritic cells. We found that MMP-2 and MMP-9 are expressed on the surface of dendritic cells from the skin, but not from other sources. They are also expressed in migrating Langerhans cells in situ. The migration of both Langerhans cells and dermal dendritic cells is inhibited by a broad spectrum inhibitor of MMPs (BB-3103), by Abs to MMP-9 and -2, and by the natural tissue inhibitors of metalloproteinases (TIMP), TIMP-1 and TIMP-2. Inhibition by anti-MMP-2 and TIMP-2 define a functional role for MMP-2 in addition to the previously described function of MMP-9. The importance of MMP-9 was emphasized using MMP-9-deficient mice in which Langerhans cell migration from skin explants was strikingly reduced. However, MMP-9 was only required for Langerhans cell migration and not maturation, since nonmigrating Langerhans cells isolated from the epidermis matured normally with regard to morphology, phenotype, and T cell stimulatory function. These data underscore the importance of MMPs, and they may be of relevance for therapeutically regulating dendritic cell migration in clinical vaccination approaches.


Immunity | 2010

Immobilized Chemokine Fields and Soluble Chemokine Gradients Cooperatively Shape Migration Patterns of Dendritic Cells

Kathrin Schumann; Tim Lämmermann; Markus Bruckner; Daniel F. Legler; Julien Polleux; Joachim P. Spatz; Gerold Schuler; Reinhold Förster; Manfred B. Lutz; Lydia Sorokin; Michael Sixt

Chemokines orchestrate immune cell trafficking by eliciting either directed or random migration and by activating integrins in order to induce cell adhesion. Analyzing dendritic cell (DC) migration, we showed that these distinct cellular responses depended on the mode of chemokine presentation within tissues. The surface-immobilized form of the chemokine CCL21, the heparan sulfate-anchoring ligand of the CC-chemokine receptor 7 (CCR7), caused random movement of DCs that was confined to the chemokine-presenting surface because it triggered integrin-mediated adhesion. Upon direct contact with CCL21, DCs truncated the anchoring residues of CCL21, thereby releasing it from the solid phase. Soluble CCL21 functionally resembles the second CCR7 ligand, CCL19, which lacks anchoring residues and forms soluble gradients. Both soluble CCR7 ligands triggered chemotactic movement, but not surface adhesion. Adhesive random migration and directional steering cooperate to produce dynamic but spatially restricted locomotion patterns closely resembling the cellular dynamics observed in secondary lymphoid organs.


European Journal of Immunology | 2009

Myeloid‐derived suppressor cell activation by combined LPS and IFN‐γ treatment impairs DC development

Verena Greifenberg; Eliana Ribechini; Susanne Rößner; Manfred B. Lutz

Myeloid‐derived suppressor cells (MDSC) and DC are major controllers of immune responses against tumors or infections. However, it remains unclear how DC development and MDSC suppressor activity both generated from myeloid precursor cells are regulated. Here, we show that the combined treatment of BM‐derived MDSC with LPS plus IFN‐γ inhibited the DC development but enhanced MDSC functions, such as NO release and T‐cell suppression. This was not observed by the single treatments in vitro. In the spleens of healthy mice, we identified two Gr‐1lowCD11bhighLy‐6ChighSSClowMo‐MDSC and Gr‐1highCD11blowPMN‐MDSC populations with suppressive potential, whereas Gr‐1highCD11bhigh neutrophils and Gr‐1lowCD11bhighSSClow eosinophils were not suppressive. Injections of LPS plus IFN‐γ expanded these populations within the spleen but not LN leading to the block of the proliferation of CD8+ T cells. At the same time, their capacity to develop into DC was impaired. Together, our data suggest that spleens of healthy mice contain two subsets of MDSC with suppressive potential. A two‐signal‐program through combined LPS and IFN‐γ treatment expands and fully activates MDSC in vitro and in vivo.


Journal of Immunology | 2001

The production of IFN-gamma by IL-12/IL-18-activated macrophages requires STAT4 signaling and is inhibited by IL-4.

Heike Schindler; Manfred B. Lutz; Martin Röllinghoff; Christian Bogdan

Macrophages release IFN-γ on combined stimulation with IL-12 and IL-18, but the signaling requirements of this process and its regulation by other cytokines are unknown. Here, we demonstrate that STAT4 is indispensable for IL-12/IL-18-induced production of IFN-γ by mouse peritoneal macrophages. Type 2 NO synthase (NOS2), which we previously found to be a prerequisite for IL-12-induced IFN-γ production in NK cells, was not required for IFN-γ production by these macrophages. IL-12 alone already induced the expression of IFN-γ mRNA, but nuclear translocation of STAT4, the release of IFN-γ protein, and the subsequent production of NO was strictly dependent on the simultaneous presence of IL-18. NF-κB, which mediates IL-18 effects in T cells, was only weakly activated by IL-12 and/or IL-18 in macrophages. Known inhibitors of macrophage functions (e.g., IL-4 and TGF-β) also suppressed macrophage IFN-γ production and the subsequent production of NOS2-derived NO. The inhibitory effect of IL-4 was paralleled by nuclear translocation of STAT6, which in EMSAs was able to bind to the same DNA oligonucleotide as STAT4. These results further define the production of IFN-γ by macrophages and point to a diversity in the signals required for IFN-γ production by various cell types.

Collaboration


Dive into the Manfred B. Lutz's collaboration.

Top Co-Authors

Avatar

Gerold Schuler

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susanne Rößner

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Alexander Steinkasserer

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Nikolaus Romani

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carsten Wiethe

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Mauritius Menges

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Susanne Rössner

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

André Gessner

University of Erlangen-Nuremberg

View shared research outputs
Researchain Logo
Decentralizing Knowledge