Network


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

Hotspot


Dive into the research topics where Lars Ohl is active.

Publication


Featured researches published by Lars Ohl.


Nature | 2002

Balanced responsiveness to chemoattractants from adjacent zones determines B-cell position

Karin Reif; Eric H. Ekland; Lars Ohl; Hideki Nakano; Martin Lipp; Reinhold Förster; Jason G. Cyster

B lymphocytes re-circulate between B-cell-rich compartments (follicles or B zones) in secondary lymphoid organs, surveying for antigen. After antigen binding, B cells move to the boundary of B and T zones to interact with T-helper cells. Despite the importance of B–T-cell interactions for the induction of antibody responses, the mechanism causing B-cell movement to the T zone has not been defined. Here we show that antigen-engaged B cells have increased expression of CCR7, the receptor for the T-zone chemokines CCL19 and CCL21, and that they exhibit increased responsiveness to both chemoattractants. In mice lacking lymphoid CCL19 and CCL21 chemokines, or with B cells that lack CCR7, antigen engagement fails to cause movement to the T zone. Using retroviral-mediated gene transfer we demonstrate that increased expression of CCR7 is sufficient to direct B cells to the T zone. Reciprocally, overexpression of CXCR5, the receptor for the B-zone chemokine CXCL13, is sufficient to overcome antigen-induced B-cell movement to the T zone. These findings define the mechanism of B-cell relocalization in response to antigen, and establish that cell position in vivo can be determined by the balance of responsiveness to chemoattractants made in separate but adjacent zones.


Journal of Experimental Medicine | 2004

Thymic T Cell Development and Progenitor Localization Depend on CCR7

Ana Misslitz; Oliver Pabst; Gabriele Hintzen; Lars Ohl; Elisabeth Kremmer; Howard T. Petrie; Reinhold Förster

T cell differentiation in the adult thymus depends on sequential interactions between lymphoid progenitors and stromal cells found in distinct regions of the cortex and medulla. Therefore, migration of T cell progenitors through distinct stromal environments seems to be a crucial process regulating differentiation and homeostasis inside the thymus. Here we show that CCR7-deficient mice are distinguished by a disturbed thymic architecture, impaired T cell development, and decreased numbers of the thymocytes. Analysis of developing double negative (CD4−CD8−) pool of wild-type thymus reveals that CCR7 expression is restricted to a CD25intCD44+ subpopulation. Correspondingly, CCR7 deficiency results in an accumulation of this population in mutant thymus. Furthermore, immunohistology shows that in CCR7-deficient mice CD25+CD44+ cells accumulate at the cortico-medullary junction, suggesting that CCR7 signaling regulates the migration of early progenitors toward the outer thymic cortex, thereby continuing differentiation. Results obtained from mixed bone marrow chimeras support this view, since the development of CCR7-deficient thymocytes is also disturbed in a morphologically intact thymus. Thus, our findings establish an essential role for CCR7 in intrathymic migration and proper T cell development.


Journal of Experimental Medicine | 2004

Chemokine Receptor CCR9 Contributes to the Localization of Plasma Cells to the Small Intestine

Oliver Pabst; Lars Ohl; Meike Wendland; Marc-André Wurbel; Elisabeth Kremmer; Bernard Malissen; Reinhold Förster

Humoral immunity in the gut-associated lymphoid tissue is characterized by the production of immunoglobulin A (IgA) by antibody-secreting plasma cells (PCs) in the lamina propria. The chemokine CCL25 is expressed by intestinal epithelial cells and is capable of inducing chemotaxis of IgA+ PCs in vitro. Using a newly generated monoclonal antibody against murine CCR9, we show that IgA+ PCs express high levels of CCR9 in the mesenteric lymph node (MLN) and Peyers patches (PPs), but down-regulate CCR9 once they are located in the small intestine. In CCR9-deficient mice, IgA+ PCs are substantially reduced in number in the lamina propria of the small intestine. In adoptive transfer experiments, CCR9-deficient IgA+ PCs show reduced migration into the small intestine compared with wild-type controls. Furthermore, CCR9 mutants fail to mount a regular IgA response to an orally administered antigen, although the architecture and cell type composition of PPs and MLN are unaffected and are functional for the generation of IgA PCs. These findings provide profound in vivo evidence that CCL25/CCR9 guides PCs into the small intestine.


Journal of Experimental Medicine | 2003

Cooperating Mechanisms of CXCR5 and CCR7 in Development and Organization of Secondary Lymphoid Organs

Lars Ohl; Golo Henning; Stefan Krautwald; Martin Lipp; Svenja Hardtke; Giinter Bernhardt; Oliver Pabst; Reinhold Förster

Homeostatic chemokines participate in the development of secondary lymphoid organs and later on in the functional organization of these tissues. The development of lymph nodes (LNs) and Peyers patches depends on the recruitment of CD3− CD4+ interleukin (IL)-7Rαhi cells to sites of future organ development. CD3− CD4+ IL-7Rαhi cells express the chemokine receptor CXCR5 and might be attracted by its ligand CXCL13, which is secreted by mesenchymal cells. Mesenchymal cells also secrete CCL19, a ligand for CCR7, yet it is not clear whether CCR7 and CCL19 are important for secondary lymphoid organ development. Analyzing CXCR5−/− CCR7−/− double deficient mice we now show that these mice lack all examined peripheral LNs suggesting a profound role for both receptors in secondary lymphoid organ development. We demonstrate that CD3− CD4+ IL-7Rαhi cells express CXCR5 as well as CCR7 indicating that both receptors cooperate during an early step of secondary lymphoid organ development. Furthermore, CXCR5−/− CCR7−/− mice display a severely disturbed architecture of mesenteric LN and spleen. Due to an impaired migration of B cells into the white pulp, CXCR5−/− CCR7−/− mice fail to develop B cell follicles but show small clusters of unorganized lymphocytes in the spleen. These data demonstrate a cooperative function of CXCR5 and CCR7 in lymphoid organ organogenesis and organization.


Journal of Immunology | 2006

Induction of Tolerance to Innocuous Inhaled Antigen Relies on a CCR7-Dependent Dendritic Cell-Mediated Antigen Transport to the Bronchial Lymph Node

Gabriele Hintzen; Lars Ohl; Maria-Luisa del Rio; Jose-Ignacio Rodriguez-Barbosa; Oliver Pabst; Jessica R. Kocks; Janet Krege; Svenja Hardtke; Reinhold Förster

Allergic airway diseases such as asthma are caused by a failure of the immune system to induce tolerance against environmental Ags. The underlying molecular and cellular mechanisms that initiate tolerance are only partly understood. In this study, we demonstrated that a CCR7-dependent migration of both CD103+ and CD103− lung dendritic cells (DC) to the bronchial lymph node (brLN) is indispensable for this process. Although inhaled Ag is amply present in the brLN of CCR7-deficient mice, T cells cannot be tolerized because of the impaired migration of Ag-carrying DC and subsequent transport of Ag from the lung to the draining lymph node. Consequently, the repeated inhalation of Ag protects wild-type but not CCR7-deficient mice from developing allergic airway diseases. Thus, the continuous DC-mediated transport of inhaled Ag to the brLN is critical for the induction of tolerance to innocuous Ags.


Journal of Experimental Medicine | 2007

Regulatory T cells interfere with the development of bronchus-associated lymphoid tissue

Jessica R. Kocks; Ana Clara Marques Davalos-Misslitz; Gabriele Hintzen; Lars Ohl; Reinhold Förster

Presence and extent of bronchus-associated lymphoid tissue (BALT) is subject to considerable variations between species and is only occasionally observed in lungs of mice. Here we demonstrate that mice deficient for the chemokine receptor CCR7 regularly develop highly organized BALT. These structures were not present at birth but were detectable from day 5 onwards. Analyzing CCR7−/−/wild-type bone marrow chimeras, we demonstrate that the development of BALT is caused by alterations of the hematopoietic system in CCR7-deficient mice. These observations together with the finding that CCR7-deficient mice posses dramatically reduced numbers of regulatory T cells (T reg cells) in the lung-draining bronchial lymph node suggest that BALT formation might be caused by disabled in situ function of T reg cells. Indeed, although adoptive transfer of wild-type T reg cells to CCR7-deficient recipients resulted in a profound reduction of BALT formation, neither naive wild-type T cells nor T reg cells from CCR7−/− donors impair BALT generation. Furthermore, we provide evidence that CCR7-deficient T reg cells, although strongly impaired in homing to peripheral lymph nodes, are fully effective in vitro. Thus our data reveal a CCR7-dependent homing of T reg cells to peripheral lymph nodes in conjunction with a role for these cells in controlling BALT formation.


Immunology | 2004

Ectopic expression of CCL19 impairs alloimmune response in mice

Stefan Krautwald; Ekkehard Ziegler; Reinhold Förster; Lars Ohl; Kerstin Amann; Ulrich Kunzendorf

Initiation of an antitumour immune response is characterized by a complex process of chemokine‐mediated cell migration and cell–cell interactions. Overexpression of chemokine CCL19 in tumour cells has been shown to result in accelerated tumour rejection under certain experimental conditions, suggesting a novel approach in the therapy of neoplastic malignancies. To investigate CCL19‐mediated modulations of cellular immune responses in vivo, we generated a chimeric CCL19‐immunoglobulin G2b (IgG2b) Fc fusion protein, which binds to the chemokine receptor CCR7 comparable to native CCL19. CCL19‐IgG2b possesses a long‐lasting potent chemotactic activity as a result of the extended half‐life of Fc fusion proteins. Stable overexpression of CCL19‐IgG2b in BALB/c‐derived J558L tumour cells fails to support tumour cell rejection following transplantation in syngeneic mice. Moreover, overexpression of CCL19‐IgG2b hinders tumour rejection in allogeneic C57BL/6 mice. This phenomenon was accompanied by a six‐fold increase of dendritic cells (DCs) isolated from CCL19‐IgG2b‐secreting tumours when compared to the number of DCs isolated from control parental J558L tumours. While mice bearing the allogeneic parental tumour showed an intense hypercellularity in the draining lymph nodes, no such response could be observed in the draining lymph nodes of mice carrying the CCL19‐IgG2b‐secreting tumour. We could demonstrate that overexpression of CCL19‐IgG2b in tumour cells retains antigen‐presenting cells in the tumour mass and prevents DCs from migrating into draining lymph nodes to present antigens and to activate T cells, resulting in an impaired immune response against the tumour.


Transplantation | 2008

Increased Transplant Arteriosclerosis in the Absence of CCR7 is Associated With Reduced Expression of Foxp3

Stephan M. Ensminger; Stephanie N. Helm; Lars Ohl; Bernd M. Spriewald; Silke Abele; M. Wollin; Kathryn J. Wood; M. Weyand; Reinhold Förster

Background. The chemokine receptor CCR7 plays a pivotal role in the homing of naïve T cells and regulatory T cells to secondary lymphoid organs and the migration of dendritic cells into afferent lymphatic vessels. Antigen presentation, T cell recruitment, and expansion of regulatory cells are crucial events in establishing and controlling chronic allograft dysfunction. In this study, we report an important role for chemokine receptor CCR7 in the development of transplant arteriosclerosis. Methods. Fully major histocompatibility complex-mismatched CBA (H2k) donor aortas were transplanted into BALB/c-CCR7−/− (H2d), BALB/c-CCR7+/− (H2d), or BALB/c-CCR7+/+ (H2d) recipients. Grafts were analyzed by histology, morphometry, and immunofluorescence on day 30 after transplantation. Intragraft cytokine mRNA production was analyzed by realtime polymerase chain reaction on day 14 after transplantation. Results. After implanting fully major histocompatibility complex-mismatched donor aortas into CCR7-deficient recipients, transplant arteriosclerosis was significantly elevated. CD4 depletion resulted in a reduction of intima proliferation in CCR7−/− recipients whereas CD8 depletion had no effect. Analysis of aortic grafts from CCR7−/− recipients revealed high numbers of infiltrating CD4+, F4/80+, and CD205+ cells. Furthermore, intragraft cytokine production showed higher levels of interleukin-4, interleukin-12, and eotaxin mRNA expression, whereas significantly lower Foxp3 mRNA expression was observed in CCR7−/− recipients. Conclusion. These data suggest that although alloantigen presentation in secondary lymphoid organs is hampered in CCR7-deficient recipients, this process may take place within the allograft itself, leading to increased formation of transplant arteriosclerosis. The decrease in Foxp3 expression despite increased in CD4+ T cell infiltration indicates a reduction in T regulatory cells possibly influencing the intensity of the graft rejection.


Transplant Immunology | 2009

Unaltered levels of transplant arteriosclerosis in the absence of the B cell homing chemokine receptor CXCR5

Stephan M. Ensminger; S. Abele-Ohl; Lars Ohl; Bernd M. Spriewald; M. Ramsperger-Gleixner; M. Weyand; Reinhold Förster

Chemokine receptors and their ligands are crucial for lymphocyte trafficking under both homeostatic and inflammatory conditions. The chemokine receptor CXCR5 controls B cell migration and the organization of B cell follicles. The aim of this study was to investigate the impact of CXCR5 on the development of transplant arteriosclerosis. Fully MHC mismatched BALB/c (H2(d)) donor aortas were transplanted into C57BL/6-CXCR5(-/-) (H2(b)), C57BL/6-CXCR5(+/-) (H2(b)) or C57BL/6-CXCR5(+/+) (H2(b)) recipients. Grafts were analysed by morphometry and immunofluorescence and intra-graft cytokine mRNA production was analysed by RT-PCR. Transplant arteriosclerosis was evident in CXCR5+/+ and CXCR5+/- mice and only mildly reduced in CXCR5-/- recipients indicating that absence of CXCR5 had no substantial effect on the development of transplant arteriosclerosis. Analysis of the cellular infiltrate of aortic grafts implanted in CXCR5-/- recipients revealed no differences in the number of T-cells, macrophages and B cells as compared to controls. Intra-graft cytokine production showed no significant changes in Th1 (IL-12) and Th2 (IL-4) cytokines as well as in TGF-beta and iNOS production. These data suggest that lack of CXCR5 expression by recipient T- and B-cells has little effect on the development of transplant arteriosclerosis.


Journal of Experimental Medicine | 2000

Follicular B Helper T Cells Express Cxc Chemokine Receptor 5, Localize to B Cell Follicles, and Support Immunoglobulin Production

Dagmar Breitfeld; Lars Ohl; Elisabeth Kremmer; Joachim W. Ellwart; Federica Sallusto; Martin Lipp; Reinhold Förster

Collaboration


Dive into the Lars Ohl's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Oliver Pabst

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Golo Henning

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

M. Weyand

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Martin Lipp

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Bernd M. Spriewald

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Elisabeth Kremmer

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge