Network


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

Hotspot


Dive into the research topics where Stephan Emmrich is active.

Publication


Featured researches published by Stephan Emmrich.


Genes & Development | 2010

miR-125b-2 is a potential oncomiR on human chromosome 21 in megakaryoblastic leukemia.

Jan-Henning Klusmann; Zhe Li; Katarina Böhmer; Aliaksandra Maroz; Mia Lee Koch; Stephan Emmrich; Frank J. Godinho; Stuart H. Orkin; Dirk Reinhardt

Children with trisomy 21/Down syndrome (DS) are at high risk to develop acute megakaryoblastic leukemia (DS-AMKL) and the related transient leukemia (DS-TL). The factors on human chromosome 21 (Hsa21) that confer this predisposing effect, especially in synergy with consistently mutated transcription factor GATA1 (GATA1s), remain poorly understood. Here, we investigated the role of Hsa21-encoded miR-125b-2, a microRNA (miRNA) overexpressed in DS-AMKL/TL, in hematopoiesis and leukemogenesis. We identified a function of miR-125b-2 in increasing proliferation and self-renewal of human and mouse megakaryocytic progenitors (MPs) and megakaryocytic/erythroid progenitors (MEPs). miR-125b-2 overexpression did not affect megakaryocytic and erythroid differentiation, but severely perturbed myeloid differentiation. The proproliferative effect of miR-125b-2 on MEPs accentuated the Gata1s mutation, whereas growth of DS-AMKL/TL cells was impaired upon miR-125b repression, suggesting synergism during leukemic transformation in GATA1s-mutated DS-AMKL/TL. Integrative transcriptome analysis of hematopoietic cells upon modulation of miR-125b expression levels uncovered a set of miR-125b target genes, including DICER1 and ST18 as direct targets. Gene Set Enrichment Analysis revealed that this target gene set is down-regulated in DS-AMKL patients highly expressing miR-125b. Thus, we propose miR-125b-2 as a positive regulator of megakaryopoiesis and an oncomiR involved in the pathogenesis of trisomy 21-associated megakaryoblastic leukemia.


Genes & Development | 2014

miR-99a/100~125b tricistrons regulate hematopoietic stem and progenitor cell homeostasis by shifting the balance between TGFβ and Wnt signaling.

Stephan Emmrich; Mareike Rasche; Jennifer Schöning; Christina Reimer; Sarva Keihani; Aliaksandra Maroz; Ying Xie; Zhe Li; Axel Schambach; Dirk Reinhardt; Jan-Henning Klusmann

Although regulation of stem cell homeostasis by microRNAs (miRNAs) is well studied, it is unclear how individual miRNAs genomically encoded within an organized polycistron can interact to induce an integrated phenotype. miR-99a/100, let-7, and miR-125b paralogs are encoded in two tricistrons on human chromosomes 11 and 21. They are highly expressed in hematopoietic stem cells (HSCs) and acute megakaryoblastic leukemia (AMKL), an aggressive form of leukemia with poor prognosis. Here, we show that miR-99a/100∼125b tricistrons are transcribed as a polycistronic message transactivated by the homeobox transcription factor HOXA10. Integrative analysis of global gene expression profiling, miRNA target prediction, and pathway architecture revealed that miR-99a/100, let-7, and miR-125b functionally converge at the combinatorial block of the transforming growth factor β (TGFβ) pathway by targeting four receptor subunits and two SMAD signaling transducers. In addition, down-regulation of tumor suppressor genes adenomatous polyposis coli (APC)/APC2 stabilizes active β-catenin and enhances Wnt signaling. By switching the balance between Wnt and TGFβ signaling, the concerted action of these tricistronic miRNAs promoted sustained expansion of murine and human HSCs in vitro or in vivo while favoring megakaryocytic differentiation. Hence, our study explains the high phylogenetic conservation of the miR-99a/100∼125b tricistrons controlling stem cell homeostasis, the deregulation of which contributes to the development of AMKL.


Molecular Cancer | 2014

LincRNAs MONC and MIR100HG act as oncogenes in acute megakaryoblastic leukemia

Stephan Emmrich; Alexandra Streltsov; Franziska Schmidt; Veera Raghavan Thangapandi; Dirk Reinhardt; Jan-Henning Klusmann

BackgroundLong non-coding RNAs (lncRNAs) are recognized as pivotal players during developmental ontogenesis and pathogenesis of cancer. The intronic microRNA (miRNA) clusters miR-99a ~ 125b-2 and miR-100 ~ 125b-1 promote progression of acute megakaryoblastic leukemia (AMKL), an aggressive form of hematologic cancers. The function of the lncRNA hostgenes MIR99AHG (alias MONC) and MIR100HG within this ncRNA ensemble remained elusive.ResultsHere we report that lncRNAs MONC and MIR100HG are highly expressed in AMKL blasts. The transcripts were mainly localized in the nucleus and their expression correlated with the corresponding miRNA clusters. Knockdown of MONC or MIR100HG impeded leukemic growth of AMKL cell lines and primary patient samples. The development of a lentiviral lncRNA vector to ectopically express lncRNAs without perturbing their secondary structure due to improper termination of the viral transcript, allowed us to study the function of MONC independent of the miRNAs in cord blood hematopoietic stem and progenitor cells (HSPCs). We could show that MONC interfered with hematopoietic lineage decisions and enhanced the proliferation of immature erythroid progenitor cells.ConclusionsOur study reveals an unprecedented function of lncRNAs MONC and MIR100HG as regulators of hematopoiesis and oncogenes in the development of myeloid leukemia.


Leukemia | 2014

miR-9 is a tumor suppressor in pediatric AML with t(8;21)

Stephan Emmrich; Jenny E. Katsman-Kuipers; K Henke; Khatib Me; Razan Jammal; F Engeland; F Dasci; Christian M. Zwaan; den Boer Ml; Lonneke J. Verboon; Jan Stary; André Baruchel; de Haas; Danen-van Oorschot Aa; Maarten Fornerod; Rob Pieters; Dirk Reinhardt; Jan-Henning Klusmann; van den Heuvel-Eibrink Mm

MicroRNAs (miRNAs) play a pivotal role in the regulation of hematopoiesis and development of leukemia. Great interest emerged in modulating miRNA expression for therapeutic purposes. In order to identify miRNAs, which specifically suppress leukemic growth of acute myeloid leukemia (AML) with t(8;21), inv(16) or mixed lineage leukemia (MLL) rearrangement by inducing differentiation, we conducted a miRNA expression profiling in a cohort of 90 cytogenetically characterized, de novo pediatric AML cases. Four miRNAs, specifically downregulated in MLL-rearranged, t(8;21) or inv(16) AMLs, were characterized by their tumor-suppressive properties in cell lines representing those respective cytogenetic groups. Among those, forced expression of miR-9 reduced leukemic growth and induced monocytic differentiation of t(8;21) AML cell lines in vitro and in vivo. The tumor-suppressive functions of miR-9 were specifically restricted to AML cell lines and primary leukemic blasts with t(8;21). On the other hand, these functions were not evident in AML blasts from patients with MLL rearrangements. We showed that miR-9 exerts its effects through the cooperation with let-7 to repress the oncogenic LIN28B/HMGA2 axis. Thus, miR-9 is a tumor suppressor-miR which acts in a stringent cell context-dependent manner.


Annals of Hematology | 2012

miRNAs can increase the efficiency of ex vivo platelet generation.

Stephan Emmrich; K Henke; Jan Hegermann; Matthias Ochs; Dirk Reinhardt; Jan-Henning Klusmann

The process of megakaryopoiesis culminates in the release of platelets, the pivotal cellular component for hemostasis and wound healing. The regulatory architecture including the modulatory role of microRNAs, which underlies megakaryocytic maturation and platelet formation, is incompletely understood, precluding the ex vivo generation of sufficient platelet numbers for transfusion medicine. We derived a highly efficient differentiation protocol to produce mature polyploid megakaryocytes and functional platelets from CD34+-hematopoietic stem and progenitor cells by comparing previously published approaches. Our megakaryocytic culture conditions using the cytokines SCF, TPO, IL-9, and IL-6 include nicotinamide and Rho-associated kinase (ROCK) inhibitor Y27632 as contextual additives. The potency of our novel megakaryocytic differentiation protocol was validated using cord blood and peripheral blood human hematopoietic stem and progenitor cells. Using this novel megakaryocytic differentiation protocol, we characterized the modulatory capacity of several miRNAs highly expressed in normal megakaryocytic cells or malignant blasts from patients with megakaryoblastic leukemia. Overexpression of candidate microRNAs was achieved by lentiviral transduction of CD34+-hematopoietic stem and progenitor cells prior to differentiation. We revealed miR-125b and miR-660 as enhancers of polyploidization, as well as platelet output of megakaryocytes. The oncogene miR-125b markedly expanded the number of megakaryocytes during in vitro culture. Conversely, the miR-23a/27a/24-2 cluster, which is highly expressed in normal megakaryocytes, blocked maturation and platelet formation. Our study on the utilization of microRNAs in conjunction with a highly efficient differentiation protocol constitutes another step towards ex vivo platelet manufacturing on a clinically relevant scale.


Oncogene | 2016

miR-139-5p controls translation in myeloid leukemia through EIF4G2

Stephan Emmrich; F Engeland; M El-Khatib; K Henke; Askar Obulkasim; J Schöning; Jenny E. Katsman-Kuipers; C. Michel Zwaan; Andreas Pich; Jan Stary; André Baruchel; V de Haas; Dirk Reinhardt; Maarten Fornerod; M.M. van den Heuvel-Eibrink; Jan-Henning Klusmann

MicroRNAs (miRNAs) are crucial components of homeostatic and developmental gene regulation. In turn, dysregulation of miRNA expression is a common feature of different types of cancer, which can be harnessed therapeutically. Here we identify miR-139-5p suppression across several cytogenetically defined acute myeloid leukemia (AML) subgroups. The promoter of mir-139 was transcriptionally silenced and could be reactivated by histone deacetylase inhibitors in a dose-dependent manner. Restoration of mir-139 expression in cell lines representing the major AML subgroups (t[8;21], inv[16], mixed lineage leukemia-rearranged and complex karyotype AML) caused cell cycle arrest and apoptosis in vitro and in xenograft mouse models in vivo. During normal hematopoiesis, mir-139 is exclusively expressed in terminally differentiated neutrophils and macrophages. Ectopic expression of mir-139 repressed proliferation of normal CD34+-hematopoietic stem and progenitor cells and perturbed myelomonocytic in vitro differentiation. Mechanistically, mir-139 exerts its effects by repressing the translation initiation factor EIF4G2, thereby reducing overall protein synthesis while specifically inducing the translation of cell cycle inhibitor p27Kip1. Knockdown of EIF4G2 recapitulated the effects of mir-139, whereas restoring EIF4G2 expression rescued the mir-139 phenotype. Moreover, elevated miR-139-5p expression is associated with a favorable outcome in a cohort of 165 pediatric patients with AML. Thus, mir-139 acts as a global tumor suppressor-miR in AML by controlling protein translation. As AML cells are dependent on high protein synthesis rates controlling the expression of mir-139 constitutes a novel path for the treatment of AML.


Nature Medicine | 2017

lncRNA MIR100HG-derived miR-100 and miR-125b mediate cetuximab resistance via Wnt/β-catenin signaling

Yuanyuan Lu; Xiaodi Zhao; Qi Liu; Cunxi Li; Ramona Graves-Deal; Zheng Cao; Bhuminder Singh; Jeffrey L. Franklin; Jing Wang; Huaying Hu; Tianying Wei; Mingli Yang; Timothy J. Yeatman; Ethan Lee; Kenyi Saito-Diaz; Scott Hinger; James G. Patton; Christine H. Chung; Stephan Emmrich; Jan-Henning Klusmann; Daiming Fan; Robert J. Coffey

De novo and acquired resistance, which are largely attributed to genetic alterations, are barriers to effective anti-epidermal-growth-factor-receptor (EGFR) therapy. To generate cetuximab-resistant cells, we exposed cetuximab-sensitive colorectal cancer cells to cetuximab in three-dimensional culture. Using whole-exome sequencing and transcriptional profiling, we found that the long non-coding RNA MIR100HG and two embedded microRNAs, miR-100 and miR-125b, were overexpressed in the absence of known genetic events linked to cetuximab resistance. MIR100HG, miR-100 and miR-125b overexpression was also observed in cetuximab-resistant colorectal cancer and head and neck squamous cell cancer cell lines and in tumors from colorectal cancer patients that progressed on cetuximab. miR-100 and miR-125b coordinately repressed five Wnt/β-catenin negative regulators, resulting in increased Wnt signaling, and Wnt inhibition in cetuximab-resistant cells restored cetuximab responsiveness. Our results describe a double-negative feedback loop between MIR100HG and the transcription factor GATA6, whereby GATA6 represses MIR100HG, but this repression is relieved by miR-125b targeting of GATA6. These findings identify a clinically actionable, epigenetic cause of cetuximab resistance.


Leukemia | 2014

GATA1s induces hyperproliferation of eosinophil precursors in Down syndrome transient leukemia

Aliaksandra Maroz; Lena Stachorski; Stephan Emmrich; Katarina Reinhardt; Jian Xu; Zhen Shao; Sebastian Käbler; Tobias Dertmann; Johann Hitzler; Irene Roberts; Paresh Vyas; Gaetan Juban; Christian Hennig; Gesine Hansen; Zhe Li; Stuart H. Orkin; Dirk Reinhardt; Jan-Henning Klusmann

Transient leukemia (TL) is evident in 5–10% of all neonates with Down syndrome (DS) and associated with N-terminal truncating GATA1 mutations (GATA1s). Here we report that TL-cell clones generate abundant eosinophils in a substantial fraction of patients. Sorted eosinophils from patients with TL and eosinophilia carried the same GATA1s mutations as sorted TL blasts, consistent with their clonal origin. TL blasts exhibited a genetic program characteristic of eosinophils and differentiated along the eosinophil lineage in vitro. Similarly, ectopic expression of Gata1s, but not Gata1, in wild-type CD34+-hematopoietic stem and progenitor cells induced hyperproliferation of eosinophil promyelocytes in vitro. Although GATA1s retained the function of GATA1 to induce eosinophil genes by occupying their promoter regions, GATA1s was impaired in its ability to repress oncogenic MYC and the pro-proliferative E2F transcription network. Chromatin Immunoprecipitation Sequencing (ChIP-seq) indicated reduced GATA1s occupancy at the MYC promoter. Knockdown of MYC, or the obligate E2F-cooperation partner DP1, rescued the GATA1s-induced hyperproliferative phenotype. In agreement, terminal eosinophil maturation was blocked in Gata1Δe2 knockin mice, exclusively expressing Gata1s, leading to accumulation of eosinophil precursors in blood and bone marrow. These data suggest a direct relationship between the N-terminal truncating mutations of GATA1 and clonal eosinophilia in DS patients.


Nature Communications | 2017

The non-coding RNA landscape of human hematopoiesis and leukemia

Adrian Schwarzer; Stephan Emmrich; Franziska Schmidt; Dominik Beck; Michelle Ng; Christina Reimer; Felix F. Adams; Sarah Grasedieck; Damian Witte; Sebastian Käbler; Jason Wong; Anushi Shah; Yizhou Huang; Razan Jammal; Aliaksandra Maroz; Mojca Jongen-Lavrencic; Axel Schambach; Florian Kuchenbauer; John E. Pimanda; Dirk Reinhardt; Dirk Heckl; Jan-Henning Klusmann

Non-coding RNAs have emerged as crucial regulators of gene expression and cell fate decisions. However, their expression patterns and regulatory functions during normal and malignant human hematopoiesis are incompletely understood. Here we present a comprehensive resource defining the non-coding RNA landscape of the human hematopoietic system. Based on highly specific non-coding RNA expression portraits per blood cell population, we identify unique fingerprint non-coding RNAs—such as LINC00173 in granulocytes—and assign these to critical regulatory circuits involved in blood homeostasis. Following the incorporation of acute myeloid leukemia samples into the landscape, we further uncover prognostically relevant non-coding RNA stem cell signatures shared between acute myeloid leukemia blasts and healthy hematopoietic stem cells. Our findings highlight the importance of the non-coding transcriptome in the formation and maintenance of the human blood hierarchy.While micro-RNAs are known regulators of haematopoiesis and leukemogenesis, the role of long non-coding RNAs is less clear. Here the authors provide a non-coding RNA expression landscape of the human hematopoietic system, highlighting their role in the formation and maintenance of the human blood hierarchy.


Journal of Clinical Oncology | 2018

Endogenous Tumor Suppressor microRNA-193b: Therapeutic and Prognostic Value in Acute Myeloid Leukemia

Raj Bhayadia; Kathrin Krowiorz; Nadine Haetscher; Razan Jammal; Stephan Emmrich; Askar Obulkasim; Jan Fiedler; Adrian Schwarzer; Arefeh Rouhi; Michael Heuser; Susanne Wingert; Sabrina Bothur; Konstanze Döhner; Tobias Mätzig; Michelle Ng; Dirk Reinhardt; Hartmut Döhner; C. Michel Zwaan; Marry van den Heuvel Eibrink; Dirk Heckl; Maarten Fornerod; Thomas Thum; R. Keith Humphries; Michael A. Rieger; Florian Kuchenbauer; Jan-Henning Klusmann

Purpose Dysregulated microRNAs are implicated in the pathogenesis and aggressiveness of acute myeloid leukemia (AML). We describe the effect of the hematopoietic stem-cell self-renewal regulating miR-193b on progression and prognosis of AML. Methods We profiled miR-193b-5p/3p expression in cytogenetically and clinically characterized de novo pediatric AML (n = 161) via quantitative real-time polymerase chain reaction and validated our findings in an independent cohort of 187 adult patients. We investigated the tumor suppressive function of miR-193b in human AML blasts, patient-derived xenografts, and miR-193b knockout mice in vitro and in vivo. Results miR-193b exerted important, endogenous, tumor-suppressive functions on the hematopoietic system. miR-193b-3p was downregulated in several cytogenetically defined subgroups of pediatric and adult AML, and low expression served as an independent indicator for poor prognosis in pediatric AML (risk ratio ± standard error, -0.56 ± 0.23; P = .016). miR-193b-3p expression improved the prognostic value of the European LeukemiaNet risk-group stratification or a 17-gene leukemic stemness score. In knockout mice, loss of miR-193b cooperated with Hoxa9/Meis1 during leukemogenesis, whereas restoring miR-193b expression impaired leukemic engraftment. Similarly, expression of miR-193b in AML blasts from patients diminished leukemic growth in vitro and in mouse xenografts. Mechanistically, miR-193b induced apoptosis and a G1/S-phase block in various human AML subgroups by targeting multiple factors of the KIT-RAS-RAF-MEK-ERK (MAPK) signaling cascade and the downstream cell cycle regulator CCND1. Conclusion The tumor-suppressive function is independent of patient age or genetics; therefore, restoring miR-193b would assure high antileukemic efficacy by blocking the entire MAPK signaling cascade while preventing the emergence of resistance mechanisms.

Collaboration


Dive into the Stephan Emmrich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dirk Heckl

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

K Henke

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Razan Jammal

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maarten Fornerod

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Zhe Li

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge