Network


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

Hotspot


Dive into the research topics where Ron-Patrick Cadeddu is active.

Publication


Featured researches published by Ron-Patrick Cadeddu.


Leukemia | 2013

Insufficient stromal support in MDS results from molecular and functional deficits of mesenchymal stromal cells

Stefanie Geyh; S. Öz; Ron-Patrick Cadeddu; Julia Fröbel; B. Brückner; Andrea Kündgen; Roland Fenk; Ingmar Bruns; Christoph Zilkens; D. Hermsen; Norbert Gattermann; Guido Kobbe; Ulrich Germing; F. Lyko; Rainer Haas; Thomas Schroeder

Ineffective hematopoiesis is a major characteristic of myelodysplastic syndromes (MDS) causing relevant morbidity and mortality. Mesenchymal stromal cells (MSC) have been shown to physiologically support hematopoiesis, but their contribution to the pathogenesis of MDS remains elusive. We show that MSC from patients across all MDS subtypes (n=106) exhibit significantly reduced growth and proliferative capacities accompanied by premature replicative senescence. Osteogenic differentiation was significantly reduced in MDS-derived MSC, indicated by cytochemical stainings and reduced expressions of Osterix and Osteocalcin. This was associated with specific methylation patterns that clearly separated MDS–MSC from healthy controls and showed a strong enrichment for biological processes associated with cellular phenotypes and transcriptional regulation. Furthermore, in MDS–MSC, we detected altered expression of key molecules involved in the interaction with hematopoietic stem and progenitor cells (HSPC), in particular Osteopontin, Jagged1, Kit-ligand and Angiopoietin as well as several chemokines. Functionally, this translated into a significantly diminished ability of MDS-derived MSC to support CD34+ HSPC in long-term culture-initiating cell assays associated with a reduced cell cycle activity. Taken together, our comprehensive analysis shows that MSC from all MDS subtypes are structurally, epigenetically and functionally altered, which leads to impaired stromal support and seems to contribute to deficient hematopoiesis in MDS.


Leukemia | 2009

The hematopoietic stem cell in chronic phase CML is characterized by a transcriptional profile resembling normal myeloid progenitor cells and reflecting loss of quiescence.

Ingmar Bruns; Czibere A; Johannes C. Fischer; Frederik Roels; Ron-Patrick Cadeddu; Buest S; D Bruennert; Huenerlituerkoglu An; Stoecklein Nh; Singh R; Zerbini Lf; Jäger M; Guido Kobbe; Norbert Gattermann; Ralf Kronenwett; Benedikt Brors; Rainer Haas

We found that composition of cell subsets within the CD34+ cell population is markedly altered in chronic phase (CP) chronic myeloid leukemia (CML). Specifically, proportions and absolute cell counts of common myeloid progenitors (CMP) and megakaryocyte–erythrocyte progenitors (MEP) are significantly greater in comparison to normal bone marrow whereas absolute numbers of hematopoietic stem cells (HSC) are equal. To understand the basis for this, we performed gene expression profiling (Affymetrix HU-133A 2.0) of the distinct CD34+ cell subsets from six patients with CP CML and five healthy donors. Euclidean distance analysis revealed a remarkable transcriptional similarity between the CML patients’ HSC and normal progenitors, especially CMP. CP CML HSC were transcriptionally more similar to their progeny than normal HSC to theirs, suggesting a more mature phenotype. Hence, the greatest differences between CP CML patients and normal donors were apparent in HSC including downregulation of genes encoding adhesion molecules, transcription factors, regulators of stem-cell fate and inhibitors of cell proliferation in CP CML. Impaired adhesive and migratory capacities were functionally corroborated by fibronectin detachment analysis and transwell assays, respectively. Based on our findings we propose a loss of quiescence of the CML HSC on detachment from the niche leading to expansion of myeloid progenitors.


Leukemia | 2013

Salvage therapy with azacitidine increases regulatory T cells in peripheral blood of patients with AML or MDS and early relapse after allogeneic blood stem cell transplantation

Thomas Schroeder; Julia Fröbel; Ron-Patrick Cadeddu; Czibere A; Dienst A; Uwe Platzbecker; Gesine Bug; Uharek L; Roland Fenk; Ulrich Germing; N Kröger; Rainer Haas; Guido Kobbe

Salvage therapy with azacitidine increases regulatory T cells in peripheral blood of patients with AML or MDS and early relapse after allogeneic blood stem cell transplantation


Leukemia | 2016

Functional inhibition of mesenchymal stromal cells in acute myeloid leukemia

Stefanie Geyh; M. Rodríguez-Paredes; Paul Jäger; Cyrus Khandanpour; Ron-Patrick Cadeddu; J. Gutekunst; Christian Matthias Wilk; Roland Fenk; Christoph Zilkens; D. Hermsen; Ulrich Germing; Guido Kobbe; F. Lyko; Rainer Haas; Thomas Schroeder

Hematopoietic insufficiency is the hallmark of acute myeloid leukemia (AML) and predisposes patients to life-threatening complications such as bleeding and infections. Addressing the contribution of mesenchymal stromal cells (MSC) to AML-induced hematopoietic failure we show that MSC from AML patients (n=64) exhibit significant growth deficiency and impaired osteogenic differentiation capacity. This was molecularly reflected by a specific methylation signature affecting pathways involved in cell differentiation, proliferation and skeletal development. In addition, we found distinct alterations of hematopoiesis-regulating factors such as Kit-ligand and Jagged1 accompanied by a significantly diminished ability to support CD34+ hematopoietic stem and progenitor cells in long-term culture-initiating cells (LTC-ICs) assays. This deficient osteogenic differentiation and insufficient stromal support was reversible and correlated with disease status as indicated by Osteocalcin serum levels and LTC-IC frequencies returning to normal values at remission. In line with this, cultivation of healthy MSC in conditioned medium from four AML cell lines resulted in decreased proliferation and osteogenic differentiation. Taken together, AML-derived MSC are molecularly and functionally altered and contribute to hematopoietic insufficiency. Inverse correlation with disease status and adoption of an AML-like phenotype after exposure to leukemic conditions suggests an instructive role of leukemic cells on bone marrow microenvironment.


Apoptosis | 2011

The non-steroidal anti-inflammatory drugs Sulindac sulfide and Diclofenac induce apoptosis and differentiation in human acute myeloid leukemia cells through an AP-1 dependent pathway

Raminder Singh; Ron-Patrick Cadeddu; Julia Fröbel; Christian Matthias Wilk; Ingmar Bruns; Luiz F. Zerbini; Tanja Prenzel; Sonja Hartwig; Daniela Brünnert; Thomas Schroeder; Stefan Lehr; Rainer Haas; Akos Czibere

Acute myeloid leukemia is a heterogeneous disease with varying genetic and molecular pathologies. Non-steroidal anti-inflammatory drugs (NSAIDs) have been proven to possess significant anti-proliferative potential in various cancer cells in vitro and in vivo. Hence, treatment with these agents can be utilized to study disease specific anti-proliferative pathways. In this study, a total number of 42 bone marrow derived CD34+ selected de novo AML patient samples and the AML cell lines THP-1 and HL-60 were treated with the NSAIDs Sulindac sulfide and Diclofenac. We analyzed viability, apoptosis, differentiation and addressed the molecular mechanisms involved. We found a consistent induction of apoptosis and to some extent an increased myeloid differentiation capacity in NSAID treated AML cells. Comprehensive protein and gene expression profiling of Diclofenac treated AML cells revealed transcriptional activation of GADD45α and its downstream MAPK/JNK pathway as well as increased protein levels of the caspase-3 precursor. This pointed towards a role of the c-Jun NH2-terminal kinase (JNK) in NSAID mediated apoptosis that we found indeed to be dependent on JNK activity as addition of a specific JNK-inhibitor abrogated apoptosis. Furthermore, the AP-1 transcription factor family members’ c-Jun, JunB and Fra-2 were transcriptionally activated in NSAID treated AML cells and re-expression of these transcription factors led to activation of GADD45α with induction of apoptosis. Mechanistically, we demonstrate that NSAIDs induce apoptosis in AML through a novel pathway involving increased expression of AP-1 heterodimers, which by itself is sufficient to induce GADD45α expression with consecutive activation of JNK and induction of apoptosis.


Molecular & Cellular Proteomics | 2013

Platelet Proteome Analysis Reveals Integrin-dependent Aggregation Defects in Patients with Myelodysplastic Syndromes

Julia Fröbel; Ron-Patrick Cadeddu; Sonja Hartwig; Ingmar Bruns; Christian Matthias Wilk; Andrea Kündgen; Johannes C. Fischer; Thomas Schroeder; Ulrich Steidl; Ulrich Germing; Stefan Lehr; Rainer Haas; Akos Czibere

Bleeding complications are a significant clinical problem in patients with myelodysplastic syndromes even at sufficient platelet counts (>50,000/μl). However, the underlying pathology of this hemorrhagic diathesis is still unknown. Here, we analyzed the platelet proteome of patients with myelodysplastic syndromes by quantitative two-dimensional difference gel electrophoresis followed by mass spectrometric protein identification. Proteins identified with lower concentrations, such as Talin-1, Vinculin, Myosin-9, Filmain-A, and Actin play critical roles in integrin αIIbβ3 signaling and thus platelet aggregation. Despite normal agonist receptor expression, calcium flux, and granule release upon activation, the activation capacity of integrin αIIbβ3 was diminished in myelodysplastic syndrome platelets. Förster resonance energy transfer analysis showed a reduced co-localization of Talin-1 to the integrins β3-subunit, which is required for receptor activation and fibrinogen binding. In addition, platelet spreading on immobilized fibrinogen was incomplete, and platelet aggregation assays confirmed a general defect in integrin-dependent platelet aggregation in patients with myelodysplastic syndromes. Our data provide novel aspects on the molecular pathology of impaired platelet function in myelodysplastic syndromes and suggest a mechanism of defective integrin αIIbβ3 signaling that may contribute to the hemorrhagic diathesis observed in these patients.


Prostaglandins & Other Lipid Mediators | 2011

Esophageal cancer proliferation is mediated by cytochrome P450 2C9 (CYP2C9).

Moritz Schmelzle; Levent Dizdar; Hanno Matthaei; Stephan Baldus; Judith Wolters; Nina Lindenlauf; Ingmar Bruns; Ron-Patrick Cadeddu; Feride Kröpil; Stefan A. Topp; Jan Schulte am Esch; Claus F. Eisenberger; Wolfram T. Knoefel; Nikolas H. Stoecklein

Cytochrome P450 epoxygenases (CYP450) have been recently shown to promote malignant progression. Here we investigated the mRNA and protein expression and potential clinical relevance of CYP2C9 in esophageal cancer. Highest expression was detected in esophageal adenocarcinoma (EAC; n=78) and adjacent esophageal mucosa (NEM; n=79). Levels of CYP2C9 in EAC and NEM were significantly higher compared to esophageal squamous cell carcinoma (ESCC; n=105). Early tumor stages and well-differentiated tumors showed a significantly higher CYP2C9 expression compared to progressed tumors. Moreover, CYP2C9 expression was correlated to high Ki-67 labeling indices in EAC and Ki-67 positive tumor cells in EAC and ESCC. Selective inhibition of CYP2C9 decreased tumor cell proliferation (KYSE30, PT1590 and OE19) in vitro, which was abolished by 11,12-epoxyeicosatrienoic acid (11,12-EET). Cell-cycle analysis using FACS revealed that inhibition of CYP2C9 leads to a G0/G1 phase cell-cycle arrest. CYP2C9 seems to be relevant for early esophageal cancer development by promoting tumor cell proliferation. Pharmacological inhibition of CYP2C9 might contribute to a more efficient therapy in CYP2C9 highly expressing esophageal cancers.


RSC Advances | 2017

Uptake dynamics of graphene quantum dots into primary human blood cells following in vitro exposure

Stefan Fasbender; Sonja Allani; Christian Wimmenauer; Ron-Patrick Cadeddu; Katharina Raba; Johannes C. Fischer; Bekir Bulat; M. Luysberg; Claus A.M. Seidel; T. Heinzel; Rainer Haas

Human leukocytes obtained from samples of leukapheresis products of three healthy donors stimulated by granulocyte colony stimulating factor (G-CSF) were exposed to graphene quantum dots. A time- and concentration dependent uptake was observed with a significantly greater uptake into monocytes and granulocytes in comparison to lymphocytes, suggesting a better incorporation ability of cells with phagocytotic properties. The uptake rates also correlate with the cell membrane area. Looking at the different lymphoid subsets a greater uptake was found into CD19+ B-, CD56+ natural killer cells and CD34+ hematopoietic stem cells (HSC) in comparison to CD4+ T- and CD8+ T cells. Independent of the cell type studied, the observed uptake dynamics is consistent with a diffusion-driven process, which allows the determination of cell-specific membrane permeabilities for the graphene quantum dots. The toxicity of the quantum dots is relatively low resulting in a 90% viability of the entire leukocyte population after 36 hours of exposure to GQDs at a concentration of 500 μg ml−1.


International Journal of Cancer | 2016

Lenalidomide consolidation treatment in patients with multiple myeloma suppresses myelopoieses but spares erythropoiesis

Christian Matthias Wilk; Niklas Heinzler; Amelie Boquoi; Ron-Patrick Cadeddu; Tobias Strapatsas; Ariane Dienst; Fatemeh Majidi; René Deenen; Ingmar Bruns; Thomas Schroeder; Karl Köhrer; Rainer Haas; Guido Kobbe; Roland Fenk

New drugs for the treatment of multiple myeloma (MM) comprise immunomodulatory substances such as lenalidomide and related compounds. While lenalidomide has found its way into first‐line treatment as well as into relapse therapy, little is known about lenalidomide effects on normal hematopoietic stem and progenitor cells (HSPCs). In this study, we investigated whether HSPCs are influenced by lenalidomide on a phenotypic, functional and gene expression level. For that purpose, samples from patients with MM were obtained who underwent equivalent first‐line treatment including induction therapy, cytotoxic stem cell mobilization and high‐dose melphalan therapy followed by autologous blood stem cell transplantation and a subsequent uniform lenalidomide consolidation treatment within a prospective clinical trial. We found that after six months of lenalidomide therapy, the number of CD34+ HSPCs decreased. Additionally, lenalidomide affects the numerical composition of hematopoietic cells in the bone marrow while it does not affect long‐term HSPC proliferation in vitro. We found a significant amplification of fetal hemoglobin (HbF) expression on a transcriptional level and can confirm a stimulated erythropoiesis on a phenotypic level. These effects were accompanied by silencing of the TGF‐β signaling pathway on the gene expression and protein level that is known to be amplified in active MM. However, these pleiotropic effects gave no evidence for mutagenic potential. In conclusion, lenalidomide does not exert long‐term effects on proliferation of HSPCs but instead promotes erythropoiesis by shifting hemoglobin expression toward HbF and by silencing the TGF‐β signaling pathway.


Clinical Hemorheology and Microcirculation | 2015

Influence of Di(2-ethylhexyl)phthalate on migration rate and differentiation of human hematopoietic stem and progenitor cells (CD34 +)

Patrick Manz; Ron-Patrick Cadeddu; Matthias Wilk; Johannes Fischer; Birgit Fritz; Rainer Haas; Folker Wenzel

INTRODUCTION Phthalates are a group of synthetic plasticizers that are ubiquitous environmental pollutants with toxic and endocrine disrupting characteristics. DEHP is the most commonly used plasticizer in the world and is still applied to stem cell transfusion bags used for storage of hematopoietic stem and progenitor cells (CD34+ HSPC), which are transferred during stem cell transplantation. Here we examined the effect of DEHP on vitality of CD34+ HSPC as well as stem cell specific properties like migration and differentiation capacity - both important for successful stem cell transplantations. MATERIAL AND METHODS CD34+ HSPC were incubated for 24 h and 72 h with DEHP concentrations ranging from 1 μg/ml to 250 μg/ml. DEHP was diluted in DMSO. Migration rate was analyzed along an SDF-1α gradient using Transwell migration inserts. Differentiation of CD34+ HSPC was investigated after two weeks in methylcellulose with colony stimulating factors. Apoptosis rate was measured via Annexin V and 7-AAD staining. RESULTS 24 h of incubation with 10 μg/ml DEHP led to a significant (p <  0.01) decrease in migration rate of CD34+ HSPC (70.70% ± 7.53% ) with a minimum migration rate of 48.33% ± 6.72% in relation to control after incubation with 100 μg/ml DEHP for 72 h. Incubation with the highest tested DEHP concentrations (50 and 100 μg/ml) significantly (p <  0.05) altered colony formation rate and cell type distribution. Apoptosis rate of CD34+ HSPC significantly (p <  0.05) increased after incubation with concentrations of 10 μg/ml DEHP for 24 h (1.46 ± 0.19) with a maximum apoptosis rate of 2.71 ± 0.66 after 24 h incubation with the highest DEHP concentration (250 μg/ml) in relation to control. CONCLUSIONS As shown, DEHP takes impact on migration rate, apoptosis rate, and differentiation of CD34+ HSPC. As these are functions with an important role in stem cell transplantations, the usage of DEHP-free stem cell transfusion bags should be considered.

Collaboration


Dive into the Ron-Patrick Cadeddu's collaboration.

Top Co-Authors

Avatar

Rainer Haas

Ludwig Maximilian University of Munich

View shared research outputs
Top Co-Authors

Avatar

Ingmar Bruns

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Guido Kobbe

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Akos Czibere

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julia Fröbel

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Roland Fenk

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Ulrich Germing

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Stefanie Geyh

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge