Network


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

Hotspot


Dive into the research topics where Florian Weisel is active.

Publication


Featured researches published by Florian Weisel.


Nature Medicine | 2008

The proteasome inhibitor bortezomib depletes plasma cells and protects mice with lupus-like disease from nephritis

Kirsten Neubert; Silke Meister; Katrin Moser; Florian Weisel; Damian Maseda; Kerstin Amann; Carsten Wiethe; Thomas H. Winkler; Joachim R. Kalden; Rudolf A. Manz; Reinhard E. Voll

Autoantibody-mediated diseases like myasthenia gravis, autoimmune hemolytic anemia and systemic lupus erythematosus represent a therapeutic challenge. In particular, long-lived plasma cells producing autoantibodies resist current therapeutic and experimental approaches. Recently, we showed that the sensitivity of myeloma cells toward proteasome inhibitors directly correlates with their immunoglobulin synthesis rates. Therefore, we hypothesized that normal plasma cells are also hypersensitive to proteasome inhibition owing to their extremely high amount of protein biosynthesis. Here we show that the proteasome inhibitor bortezomib, which is approved for the treatment of multiple myeloma, eliminates both short- and long-lived plasma cells by activation of the terminal unfolded protein response. Treatment with bortezomib depleted plasma cells producing antibodies to double-stranded DNA, eliminated autoantibody production, ameliorated glomerulonephritis and prolonged survival of two mouse strains with lupus-like disease, NZB/W F1 and MRL/lpr mice. Hence, the elimination of autoreactive plasma cells by proteasome inhibitors might represent a new treatment strategy for antibody-mediated diseases.


Immunological Reviews | 2012

Germinal center selection and the development of memory B and plasma cells.

Mark J. Shlomchik; Florian Weisel

Summary:  A hallmark of adaptive immune responses is the generation of long‐lived protection after primary exposure to a pathogen. In humoral responses, this protection stems from a combination of sustained antibody titers and long‐lived memory B cells (MBCs), with the former deriving from long‐lived plasma cells (PCs). Both types of cell are thought to primarily derive from the germinal center (GC), a unique structure that forms during the immune response to many types of antigenic stimuli. GCs are seeded by antigen‐specific B and T cells that were previously activated in the early stages of the response. The GC does not directly or immediately generate effector function; rather, it is a site of intense B‐cell proliferation and cell death. GC B cells undergo both somatic hypermutation and isotype switch, and a Darwinian process very efficiently selects B cells with higher fitness for survival and expansion. GC B cells adopt a unique activation and transcriptional state, and the cells become poised to differentiate to either MBCs or PCs. Despite this general understanding of the events in the GC, the mechanisms that control both affinity selection as well as differentiation have not been well worked out. In this review, we address what is known about what determines whether GC B cells become MBCs or PCs. This is discussed in the broader context of the origins of both cell types, whether from the GC or potentially other sources. We present a model encompassing recent data from several laboratories including our own that suggests that the GC undergoes a temporal switch that alters the nature of its output from MBCs to PCs as the response progresses. We will discuss B‐cell receptor signaling in the GC as it relates to potential mechanisms for affinity‐based selection during the reaction.


Nature Immunology | 2007

Siglec-G is a B1 cell-inhibitory receptor that controls expansion and calcium signaling of the B1 cell population.

Anja Hoffmann; Sheena Kerr; Julia Jellusova; Jiquan Zhang; Florian Weisel; Ute Wellmann; Thomas H. Winkler; Burkhard Kneitz; Paul R. Crocker; Lars Nitschke

B1 cells are an important cell population for the production of natural antibodies and for antibacterial immunoglobulin responses. Here we identified the mouse protein Siglec-G as a B1 cell inhibitory receptor. Siglec-G was expressed in a B cell–restricted way, with large amounts present in B1 cells. When overexpressed, Siglec-G inhibited B cell receptor–mediated calcium signaling. Siglec-G-deficient mice had massive expansion of the B1a cell population, which began early in development and was B cell intrinsic. Siglec-G-deficient mice had higher titers of natural IgM antibodies but not a higher penetrance of IgG autoantibodies. Siglec-G-deficient B1 cells showed a strongly enhanced calcium signaling. Our results demonstrate that Siglec-G-dependent negative regulation exists in B1 cells, which may explain the naturally muted signaling response of B1 cells.


Nature Immunology | 2014

CD80 and PD-L2 define functionally distinct memory B cell subsets that are independent of antibody isotype

Griselda Zuccarino-Catania; Saheli Sadanand; Florian Weisel; Mary M. Tomayko; Hailong Meng; Steven H. Kleinstein; Kim L. Good-Jacobson; Mark J. Shlomchik

Memory B cells (MBCs) are long-lived sources of rapid, isotype-switched secondary antibody-forming cell (AFC) responses. Whether MBCs homogeneously retain the ability to self-renew and terminally differentiate or if these functions are compartmentalized into MBC subsets has remained unclear. It has been suggested that antibody isotype controls MBC differentiation upon restimulation. Here we demonstrate that subcategorizing MBCs on the basis of their expression of CD80 and PD-L2, independently of isotype, identified MBC subsets with distinct functions upon rechallenge. CD80+PD-L2+ MBCs differentiated rapidly into AFCs but did not generate germinal centers (GCs); conversely, CD80−PD-L2− MBCs generated few early AFCs but robustly seeded GCs. The gene-expression patterns of the subsets supported both the identity and function of these distinct MBC types. Hence, the differentiation and regeneration of MBCs are compartmentalized.


Immunity | 2016

A Temporal Switch in the Germinal Center Determines Differential Output of Memory B and Plasma Cells

Florian Weisel; Griselda Zuccarino-Catania; Maria Chikina; Mark J. Shlomchik

There is little insight into or agreement about the signals that control differentiation of memory B cells (MBCs) and long-lived plasma cells (LLPCs). By performing BrdU pulse-labeling studies, we found that MBC formation preceded the formation of LLPCs in an adoptive transfer immunization system, which allowed for a synchronized Ag-specific response with homogeneous Ag-receptor, yet at natural precursor frequencies. We confirmed these observations in wild-type (WT) mice and extended them with germinal center (GC) disruption experiments and variable region gene sequencing. We thus show that the GC response undergoes a temporal switch in its output as it matures, revealing that the reaction engenders both MBC subsets with different immune effector function and, ultimately, LLPCs at largely separate points in time. These data demonstrate the kinetics of the formation of the cells that provide stable humoral immunity and therefore have implications for autoimmunity, for vaccine development, and for understanding long-term pathogen resistance.


PLOS Pathogens | 2011

B Cell Repertoire Analysis Identifies New Antigenic Domains on Glycoprotein B of Human Cytomegalovirus which Are Target of Neutralizing Antibodies

Sonja Pötzsch; Nadja Spindler; Anna-Katharina Wiegers; Tanja Fisch; Pia Rücker; Heinrich Sticht; Nina Grieb; Tina Baroti; Florian Weisel; Thomas Stamminger; Luis Martin-Parras; Michael Mach; Thomas H. Winkler

Human cytomegalovirus (HCMV), a herpesvirus, is a ubiquitously distributed pathogen that causes severe disease in immunosuppressed patients and infected newborns. Efforts are underway to prepare effective subunit vaccines and therapies including antiviral antibodies. However, current vaccine efforts are hampered by the lack of information on protective immune responses against HCMV. Characterizing the B-cell response in healthy infected individuals could aid in the design of optimal vaccines and therapeutic antibodies. To address this problem, we determined, for the first time, the B-cell repertoire against glycoprotein B (gB) of HCMV in different healthy HCMV seropositive individuals in an unbiased fashion. HCMV gB represents a dominant viral antigenic determinant for induction of neutralizing antibodies during infection and is also a component in several experimental HCMV vaccines currently being tested in humans. Our findings have revealed that the vast majority (>90%) of gB-specific antibodies secreted from B-cell clones do not have virus neutralizing activity. Most neutralizing antibodies were found to bind to epitopes not located within the previously characterized antigenic domains (AD) of gB. To map the target structures of these neutralizing antibodies, we generated a 3D model of HCMV gB and used it to identify surface exposed protein domains. Two protein domains were found to be targeted by the majority of neutralizing antibodies. Domain I, located between amino acids (aa) 133–343 of gB and domain II, a discontinuous domain, built from residues 121–132 and 344–438. Analysis of a larger panel of human sera from HCMV seropositive individuals revealed positivity rates of >50% against domain I and >90% against domain II, respectively. In accordance with previous nomenclature the domains were designated AD-4 (Dom II) and AD-5 (Dom I), respectively. Collectively, these data will contribute to optimal vaccine design and development of antibodies effective in passive immunization.


Journal of Immunology | 2010

Unique Requirements for Reactivation of Virus-Specific Memory B Lymphocytes

Florian Weisel; Uwe Appelt; Andrea Schneider; Jasmin U. Horlitz; Nico van Rooijen; Heinrich Körner; Michael Mach; Thomas H. Winkler

Memory B cells (MBCs) are rapidly activated upon Ag re-exposure in vivo, but the precise requirements for this process are still elusive. To address these requirements, T cell-independent reactivation of MBCs against virus-like particles was analyzed. As few as 25 MBCs are sufficient for a measurable Ab response after adoptive transfer. We found that MBCs were reactivated upon antigenic challenge to normal levels after depletion of macrophages, CD11c+ dendritic cells, and matured follicular dendritic cells. Furthermore, MBC responses were possible in TNF/lymphotoxin α double-deficient mice after partial normalization of lymphoid architecture by means of long-term reconstitution with wild-type bone marrow. Activation did not occur when chimeric mice, which still lack all lymph nodes and Peyer’s patches, were splenectomized prior to MBC transfer. Together with our finding that MBC responses are weak when Ag was administered within minutes after adoptive MBC transfer, these results strongly suggest that MBCs have to occupy specific niches within secondary lymphoid tissue to become fully Ag-responsive. We provide clear evidence that MBCs are not preferentially resident within the splenic marginal zones and show that impaired homing to lymphoid follicles resulted in significantly diminished activation, suggesting that reactivation of MBCs occurred inside lymphoid follicles. Furthermore, comparison of virus-specific MBC T cell-independent reactivation versus primary T cell-independent type II B cell activation revealed unique requirements of MBC activation.


Annual Review of Immunology | 2017

Memory B Cells of Mice and Humans

Florian Weisel; Mark J. Shlomchik

We comprehensively review memory B cells (MBCs), covering the definition of MBCs and their identities and subsets, how MBCs are generated, where they are localized, how they are maintained, and how they are reactivated. Whereas naive B cells adopt multiple fates upon stimulation, MBCs are more restricted in their responses. Evolving work reveals that the MBC compartment in mice and humans consists of distinct subpopulations with differing effector functions. We discuss the various approaches to define subsets and subset-specific roles. A major theme is the need to both deliver faster effector function upon reexposure and readapt to antigenically variant pathogens while avoiding burnout, which would be the result if all MBCs generated only terminal effector function. We discuss cell-intrinsic differences in gene expression and signaling that underlie differences in function between MBCs and naive B cells and among MBC subsets and how this leads to memory responses.


Journal of Immunology | 2012

JAB1 Is Essential for B Cell Development and Germinal Center Formation and Inversely Regulates Fas Ligand and Bcl6 Expression

Selina Sitte; Joachim Gläsner; Julia Jellusova; Florian Weisel; Martina Panattoni; Ruggero Pardi; André Gessner

Jun activation domain-binding protein 1 (JAB1) regulates ubiquitin-dependent protein degradation by deneddylation of cullin-based ubiquitin ligases and, therefore, plays a central role in regulating proliferation and apoptosis. Because these processes are decisive for B cell development, we investigated JAB1 functions in B cells by establishing a mouse strain with a B cell-specific JAB1 deletion. We show that JAB1 is essential for early B cell development, because the ablation of JAB1 expression blocks B cell development between the pro-B and pre-B cell stages. Furthermore, JAB1 deletion leads to aberrant expression of the apoptosis-triggering protein Fas ligand in pro-B cells. Concomitant B cell-specific overexpression of the antiapoptotic protein Bcl2 partially reverses the block in B cell development; rescued JAB1-deficient B cells reach the periphery and produce protective class-switched Abs after Borrelia burgdorferi infection. Interestingly, B cell-rescued mice exhibit no germinal centers but a striking extrafollicular plasma cell accumulation. In addition, JAB1 is essential for Bcl6 expression, a transcriptional repressor required for germinal center formation. These findings identify JAB1 as an important factor in checkpoint control during early B cell development, as well as in fate decisions in mature Ag-primed B cells.


The EMBO Journal | 2011

A hypomorphic IgH-chain allele affects development of B-cell subsets and favours receptor editing

Sven Brenner; Diana Drewel; Thomas Steinbart; Florian Weisel; Eric Härtel; Sonja Pötzsch; Heike Welzel; Andreas Brandl; Philipp Yu; Geert Mudde; Astrid Schweizer; Lars Nitschke; Thomas H. Winkler

The quality and quantity of BCR signals impact on cell fate decisions of B lymphocytes. Here, we describe novel gene‐targeted mice, which in the context of normal VDJ recombination show hypomorphic expression of immunoglobulin μ heavy chain (μHC) mRNA levels and hence lower pre‐BCR and BCR levels. Hypomorphic expression of μHC leads to augmented selection processes at all stages of B‐cell development, noticeably at the expansion of pre‐B cells, the positive selection of immature B lymphocytes in the bone marrow and the selection of the follicular (FO), marginal zone (MZ) and B1 B‐lymphocyte compartment in peripheral lymphoid organs. Immature as well as mature FO and MZ B lymphocytes in the peripheral lymphoid organs express lower levels of the receptor for B‐cell activating factor (BAFF). In addition, hypomorphic expression of the BCR favours receptor editing. Together, our results highlight the critical importance of pre‐BCR and BCR receptor levels for the normal development of B‐lymphocyte subpopulations in the context of intact VDJ recombination and a diverse antibody repertoire.

Collaboration


Dive into the Florian Weisel's collaboration.

Top Co-Authors

Avatar

Thomas H. Winkler

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Mach

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Ute Wellmann

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Lars Nitschke

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Sonja Pötzsch

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Uwe Appelt

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea Schneider

University of Erlangen-Nuremberg

View shared research outputs
Researchain Logo
Decentralizing Knowledge