Network


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

Hotspot


Dive into the research topics where Bernhard Schiedlmeier is active.

Publication


Featured researches published by Bernhard Schiedlmeier.


Molecular Therapy | 2008

Self-inactivating gammaretroviral vectors for gene therapy of x-linked severe combined immunodeficiency

Susannah I Thornhill; Axel Schambach; Steven J. Howe; Meera Ulaganathan; Elke Grassman; David A. Williams; Bernhard Schiedlmeier; Nj Sebire; H. Bobby Gaspar; Christine Kinnon; Christopher Baum; Adrian J. Thrasher

Gene therapy for X-linked severe combined immunodeficiency (SCID-X1) has proven highly effective for long-term restoration of immunity in human subjects. However, the development of lymphoproliferative complications due to dysregulated proto-oncogene expression has underlined the necessity for developing safer vector systems. To reduce the potential for insertional mutagenesis, we have evaluated the efficacy of self-inactivating (SIN) gammaretroviral vectors in cellular and in vivo models of SCID-X1. Vectors incorporating an internal human elongation factor-1alpha regulatory element were capable of fully restoring the lymphoid differentiation potential of gammac-deficient lineage negative cells. Multilineage lymphoid reconstitution of a murine model was achieved at a similar level to that achieved by a conventional long-terminal repeat (LTR)-regulated vector used in previous clinical trials. Functional proliferative responses to mitogenic stimuli were also restored, and serum immunoglobulin levels were normalized. The reduced mutagenic potential conferred by SIN vector configurations and alternative non-LTR-based regulatory elements, together with proven efficacy in correction of cellular defects provides an important platform for development of the next phase of clinical trials for SCID-X1.


Proceedings of the National Academy of Sciences of the United States of America | 2007

HOXB4's road map to stem cell expansion

Bernhard Schiedlmeier; Ana Cristina Santos; Ana C.F. Ribeiro; Natalia Moncaut; Dietrich Lesinski; Herbert Auer; Karl Kornacker; Wolfram Ostertag; Christopher Baum; Moisés Mallo; Hannes Klump

Homeodomain-containing transcription factors are important regulators of stem cell behavior. HOXB4 mediates expansion of adult and embryo-derived hematopoietic stem cells (HSCs) when expressed ectopically. To define the underlying molecular mechanisms, we performed gene expression profiling in combination with subsequent functional analysis with enriched adult HSCs and embryonic derivatives expressing inducible HOXB4. Thereby, we identified a set of overlapping genes that likely represent “universal” targets of HOXB4. A substantial number of loci are involved in signaling pathways important for controlling self-renewal, maintenance, and differentiation of stem cells. Functional assays performed on selected pathways confirmed the biological coherence of the array results. HOXB4 activity protected adult HSCs from the detrimental effects mediated by the proinflammatory cytokine TNF-α. This protection likely contributes to the competitive repopulation advantage of HOXB4-expressing HSCs observed in vivo. The concept of TNF-α inhibition may also prove beneficial for patients undergoing bone marrow transplantation. Furthermore, we demonstrate that HOXB4 activity and FGF signaling are intertwined. HOXB4-mediated expansion of adult and ES cell-derived HSCs was enhanced by specific and complete inhibition of FGF receptors. In contrast, the expanding activity of HOXB4 on hematopoietic progenitors in day 4–6 embryoid bodies was blunted in the presence of basic FGF (FGF2), indicating a dominant negative effect of FGF signaling on the earliest hematopoietic cells. In summary, our results strongly suggest that HOXB4 modulates the response of HSCs to multiple extrinsic signals in a concerted manner, thereby shifting the balance toward stem cell self-renewal.


Molecular Therapy | 2009

Cell-intrinsic and Vector-related Properties Cooperate to Determine the Incidence and Consequences of Insertional Mutagenesis

Olga Kustikova; Bernhard Schiedlmeier; Martijn H. Brugman; Maike Stahlhut; Stefan Bartels; Zhixiong Li; Christopher Baum

In gene therapeutic approaches targeting hematopoietic cells, insertional mutagenesis may provoke clonal dominance with potential progress to overt leukemia. To investigate the contribution of cell-intrinsic features and determine the frequency of insertional proto-oncogene activation, we sorted hematopoietic subpopulations before transduction with replication-deficient gamma-retroviral vectors and studied the clonal repertoire in transplanted C57BL/6J mice. Progressive clonal dominance only developed in the progeny of populations with intrinsic stem cell potential, where expanding clones with insertional upregulation of proto-oncogenes such as Evi1 were retrieved with a frequency of approximately 10(-4). Longitudinal studies by high-throughput sequencing and locus-specific quantitative PCR showed clones with >50-fold expansion between weeks 5 and 31 after transplantation. In contrast, insertional events in proto-oncogenes did not endow the progeny of multipotent or myeloid-restricted progenitors with the potential for clonal dominance (risk <10(-6)). Transducing sorted hematopoietic stem cells (HSCs) with self-inactivating (SIN) lentiviral vectors in short-term cultures improved chimerism, and although clonal dominance developed, there was no evidence for insertional events in the vicinity of proto-oncogenes as the underlying cause. We conclude that cell-intrinsic properties cooperate with vector-related features to determine the incidence and consequences of insertional mutagenesis. Furthermore, our study offers perspectives for refinement of animal experiments in the assessment of vector-related genotoxicity.


Annals of the New York Academy of Sciences | 2005

Control of Self-Renewal and Differentiation of Hematopoietic Stem Cells: HOXB4 on the Threshold

Hannes Klump; Bernhard Schiedlmeier; Christopher Baum

Abstract: The homeodomain transcription factor HOXB4 is one of the most attractive tools to expand hematopoietic stem cells in vitro and in vivo and to promote the formation of hematopoietic cells from in vitro differentiated embryonic stem cells. However, the expression levels compatible with the favorable effect of enhanced self‐renewal without perturbing differentiation, in vivo, remain to be determined. In this paper, we discuss the necessity to define the “therapeutic width” of HOXB4 expression, based on observations from our lab and others that demonstrate that ectopic HOXB4 expression leads to a concentration‐dependent perturbation of lineage differentiation of mouse and human hematopoietic cells. In summary, the combined results argue in favor of the existence of certain threshold levels for HOXB4 activity that control the differentiation and self‐renewal behavior of hematopoietic stem and progenitor cells. Indeed, existing evidence suggests that dosage effects of ectopically expressed transcription factors may be more the rule than an exception.


Leukemia | 2007

Remarkable leukemogenic potency and quality of a constitutively active neurotrophin receptor, ΔTrkA

J Meyer; M Rhein; Bernhard Schiedlmeier; Olga Kustikova; Cornelia Rudolph; K Kamino; Thomas Neumann; Min Yang; A Wahlers; Boris Fehse; G W Reuther; Brigitte Schlegelberger; Arnold Ganser; Christopher Baum; Zhixiong Li

Neurotrophins and their receptors play a key role in neurogenesis and survival. However, we and others have recently obtained evidence for a potential involvement of this receptor system in leukemia. To investigate mechanisms underlying the leukemogenic potential of activated neurotrophin receptor signaling, we analyzed in vivo leukemogenesis mediated by ΔTrkA, a mutant of TRKA (tropomyosin-related kinase A) isolated from a patient with acute myeloid leukemia (AML). Retroviral expression of ΔTrkA in myeloid 32D cells induced AML in syngeneic C3H/Hej mice (n=11/11, latency ∼4 weeks). C57Bl/6J mice transplanted with ΔTrkA-transduced primary lineage negative (Lin−) bone marrow cells died of a transient polyclonal AML (n=7/15, latency of <12 days). Serial transplantation of AML cells did not re-induce this disease but rather acute lymphoblastic leukemia (ALL, latency >78 days). All primary recipients surviving the early AML developed clonal ALL or myeloid leukemia (latency >72 days) that required additional genetic lesions. PI3K and mTOR-raptor were identified as the crucial mediators of leukemic transformation, whereas STAT and MAP kinase signaling pathways were not activated. Thus, our findings reveal potent and unique transforming properties of altered neurotrophin receptor signaling in leukemogenesis, and encourage further analyses of neurotrophin receptors and downstream signaling events in hematological malignancies.


Leukemia | 2002

Persisting multilineage transgene expression in the clonal progeny of a hematopoietic stem cell

Zhixiong Li; Boris Fehse; Bernhard Schiedlmeier; Jochen Düllmann; Oliver Frank; Axel R. Zander; Wolfram Ostertag; Christopher Baum

Many applications of hematopoietic gene therapy require selection for clones with active transgene expression. However, it was unclear whether the clonal progeny of a retrovirally transduced hematopoietic stem cell would be capable of maintaining transgene expression through serial repopulation and multilineage differentiation. Such investigations require simultaneous analyses of clonality, multilineage activity and transgene copy numbers. Using a mouse model, the present study demonstrates that a single hematopoietic stem cell expressing a marker gene from one or two insertions of a simple retroviral vector actively maintains multilineage transgene expression in the vast majority (80–99%) of bone marrow and peripheral blood cells. Gene expression persisted through serial transplantations for at least 97 weeks post gene transfer and was observed in the lymphoid (B, T and NK cells), myeloid (CD11b+, Gr-1+), erythroid (Ter119+, mature red blood cells) and megakaryocytic (as indicated by platelets) progeny. Therefore, a single immunoselection for hematopoietic stem cells expressing the transgene in vivo was sufficient to establish a completely chimeric hematopoiesis. These observations imply that the retroviral vectors used in this study contain cis-elements that mediate expression through massive clonal expansion and multilineage differentiation, provided the insertion occurred in genetic loci permissive for expression in hematopoietic stem cells.


Blood | 2009

Ectopic HOXB4 overcomes the inhibitory effect of tumor necrosis factor-α on Fanconi anemia hematopoietic stem and progenitor cells

Michael D. Milsom; Bernhard Schiedlmeier; Jeff Bailey; Mi-Ok Kim; Dandan Li; Michael Jansen; Abdullah Mahmood Ali; Michelle Kirby; Christopher Baum; Leslie J. Fairbairn; David A. Williams

Ectopic delivery of HOXB4 elicits the expansion of engrafting hematopoietic stem cells (HSCs). We hypothesized that inhibition of tumor necrosis factor-alpha (TNF-alpha) signaling may be central to the self-renewal signature of HOXB4. Because HSCs derived from Fanconi anemia (FA) knockout mice are hypersensitive to TNF-alpha, we studied Fancc(-/-) HSCs to determine the physiologic effects of HOXB4 on TNF-alpha sensitivity and the relationship of these effects to the engraftment defect of FA HSCs. Overexpression of HOXB4 reversed the in vitro hypersensitivity to TNF-alpha of Fancc(-/-) HSCs and progenitors (P) and partially rescued the engraftment defect of these cells. Coexpression of HOXB4 and the correcting FA-C protein resulted in full correction compared with wild-type (WT) HSCs. Ectopic expression of HOXB4 resulted in a reduction in both apoptosis and reactive oxygen species in Fancc(-/-) but not WT HSC/P. HOXB4 overexpression was also associated with a significant reduction in surface expression of TNF-alpha receptors on Fancc(-/-) HSC/P. Finally, enhanced engraftment was seen even when HOXB4 was expressed in a time-limited fashion during in vivo reconstitution. Thus, the HOXB4 engraftment signature may be related to its effects on TNF-alpha signaling, and this pathway may be a molecular target for timed pharmacologic manipulation of HSC during reconstitution.


Cell Cycle | 2006

HOXB4 Inhibits Cell Growth in a Dose-Dependent Manner and Sensitizes Cells Towards Extrinsic Cues

Elke Will; Daniel Speidel; Zheng Wang; Gabriel Ghiaur; Andreas Rimek; Bernhard Schiedlmeier; David A. Williams; Christopher Baum; Wolfram Ostertag; Hannes Klump

Ectopic expression of the homeodomain transcription factor HOXB4 expandshematopoietic stem and progenitor cells in vivo and in vitro, making HOXB4 a highlyinteresting candidate for therapeutic stem cell expansion. However, when expressedat high levels, HOXB4 concomitantly perturbs differentiation and thus likelypredisposes the manipulated cells for leukemogenesis. We therefore asked whetherthe expression level of HOXB4 may be a critical parameter that influences the growthand transformation properties of transduced cells. Using a set of retroviral vectorswhich covered a 40-fold range of expression levels, we studied the consequences ofHOXB4 expression at different levels in the well established Rat-1 fibroblast cellsystem. HOXB4 transformed Rat-1 fibroblasts beyond a certain threshold level ofexpression. Further escalation of HOXB4 expression, however, did not enhancetransformation. Nevertheless, HOXB4 mediated a dose dependent anti-proliferativeeffect on Rat-1 and NIH3T3 fibroblasts. This effect was aggravated under reducedserum concentrations and was, at least partially, due to an enhanced sensitivity ofHOXB4 overexpressing cells to induction of apoptosis. Based on these results wepropose that HOXB4 affects cell growth in a dose-dependent manner by sensitizingcells towards extrinsic signals.


Gene Therapy | 2006

Towards hematopoietic stem cell-mediated protection against infection with human immunodeficiency virus

Axel Schambach; Bernhard Schiedlmeier; Klaus Kühlcke; Monique M.A. Verstegen; Geoffrey P. Margison; Zhiyuan Li; Kenji Kamino; Jens Bohne; Alexander Alexandrov; Felix Hermann; D. Von Laer; Christopher Baum

The failure of pharmacological approaches to cure infection with the human immunodeficiency virus (HIV) has renewed the interest in gene-based therapies. Among the various strategies that are currently explored, the blockade of HIV entry into susceptible T cells and macrophages promises to be the most powerful intervention. For long-term protection of both of these lineages, genetic modification of hematopoietic stem cells (HSCs) would be required. Here, we tested whether HSCs and their progeny can be modified to express therapeutic levels of M87o, a gammaretroviral vector encoding an artificial transmembrane molecule that blocks fusion-mediated uptake of HIV. In serial murine bone marrow transplantations, efficient and multilineage expression of M87o was observed for more than 1 year (range 37–75% of mononuclear cells), without signs of toxicity related to the transmembrane molecule. To allow enrichment of M87o-modified HSCs after transplant, we constructed vectors coexpressing the P140K mutant of O6-methylguanine-DNA-methyltransferase (MGMT-P140K). This clinically relevant selection marker mediates a survival advantage in HSCs if exposed to combinations of methylguanine-methyltransferase (MGMT) inhibitors and alkylating agents. A bicistronic vector mediated sufficient expression of both M87o and MGMT to confer a selective survival advantage in the presence of HIV and alkylating agents, respectively. These data encourage further investigations in large animal models and clinical trials.


Cancer Gene Therapy | 2000

Differential expression of a recombinant adeno-associated virus 2 vector in human CD34 + cells and breast cancer cells

Marlon R. Veldwijk; Stefan Fruehauf; Bernhard Schiedlmeier; Juergen Kleinschmidt; W. Jens Zeller

The use of autologous hematopoietic stem cell (HSC) grafts after high-dose chemotherapy protocols may be hampered by contamination of the grafts with tumor cells. Because epithelial cells seem to be the natural hosts of adeno-associated virus 2 (AAV-2), we speculated that epithelial tumor cells in HSC grafts might be selective targets for AAV-2-based vectors. To test this hypothesis, the breast cancer cell lines T47D and MCF-7 were infected with a recombinant AAV-2 vector expressing the green fluorescent protein (GFP) gene; in addition, human CD34+ mobilized peripheral progenitor cells were infected with the same vector. At a multiplicity of infection of 100, only 1.39% ± 0.51% CD34+ cells expressed the GFP gene whereas, 36.06% ± 6.53% of the infected T47D cells and 41.52% ± 3.16% of the infected MCF-7 cells expressed the transduced GFP gene. After further optimizing the transduction procedure by using higher multiplicities of infection (100–500) and preincubation of samples with the tyrosine kinase inhibitor genistein, up to 82.52% and 85.35% GFP+ T47D and MCF-7 cells, respectively, were observed. The GFP fluorescence intensity in transduced mammary tumor cells was up to 3 logs higher than that of transduced CD34+ cells. The differential expression of recombinant AAV-2 vectors in hematopoietic and epithelial tumor cells warrants further research with this vector system, including the use of suicide genes for the purging of autologous HSC grafts.

Collaboration


Dive into the Bernhard Schiedlmeier's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hannes Klump

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhixiong Li

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Niels Heinz

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Ute Modlich

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge