Network


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

Hotspot


Dive into the research topics where Matthias Bartenstein is active.

Publication


Featured researches published by Matthias Bartenstein.


Gastroenterology | 2013

Hypomethylation of Noncoding DNA Regions and Overexpression of the Long Noncoding RNA, AFAP1-AS1, in Barrett’s Esophagus and Esophageal Adenocarcinoma

Wenjing Wu; Tushar D. Bhagat; Xue Yang; Jee Hoon Song; Yulan Cheng; Rachana Agarwal; John M. Abraham; Sariat Ibrahim; Matthias Bartenstein; Zulfiqar Hussain; Masako Suzuki; Yiting Yu; Wei Chen; Charis Eng; John M. Greally; Amit Verma; Stephen J. Meltzer

BACKGROUND & AIMS Alterations in methylation of protein-coding genes are associated with Barretts esophagus (BE) and esophageal adenocarcinoma (EAC). Dysregulation of noncoding RNAs occurs during carcinogenesis but has never been studied in BE or EAC. We applied high-resolution methylome analysis to identify changes at genomic regions that encode noncoding RNAs in BE and EAC. METHODS We analyzed methylation of 1.8 million CpG sites using massively parallel sequencing-based HELP tagging in matched EAC, BE, and normal esophageal tissues. We also analyzed human EAC (OE33, SKGT4, and FLO-1) and normal (HEEpic) esophageal cells. RESULTS BE and EAC exhibited genome-wide hypomethylation, significantly affecting intragenic and repetitive genomic elements as well as noncoding regions. These methylation changes targeted small and long noncoding regions, discriminating normal from matched BE or EAC tissues. One long noncoding RNA, AFAP1-AS1, was extremely hypomethylated and overexpressed in BE and EAC tissues and EAC cells. Its silencing by small interfering RNA inhibited proliferation and colony-forming ability, induced apoptosis, and reduced EAC cell migration and invasion without altering the expression of its protein-coding counterpart, AFAP1. CONCLUSIONS BE and EAC exhibit reduced methylation that includes noncoding regions. Methylation of the long noncoding RNA AFAP1-AS1 is reduced in BE and EAC, and its expression inhibits cancer-related biologic functions of EAC cells.


Blood | 2015

IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells

Carolina Schinke; Orsolya Giricz; Weijuan Li; Aditi Shastri; Shanisha Gordon; Laura Barreyro; Laura Barreryo; Tushar D. Bhagat; Sanchari Bhattacharyya; Nandini Ramachandra; Matthias Bartenstein; Andrea Pellagatti; Jacqueline Boultwood; Amittha Wickrema; Yiting Yu; Britta Will; Sheng Wei; Ulrich Steidl; Amit Verma

Acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are associated with disease-initiating stem cells that are not eliminated by conventional therapies. Novel therapeutic targets against preleukemic stem cells need to be identified for potentially curative strategies. We conducted parallel transcriptional analysis of highly fractionated stem and progenitor populations in MDS, AML, and control samples and found interleukin 8 (IL8) to be consistently overexpressed in patient samples. The receptor for IL8, CXCR2, was also significantly increased in MDS CD34(+) cells from a large clinical cohort and was predictive of increased transfusion dependence. High CXCR2 expression was also an adverse prognostic factor in The Cancer Genome Atlas AML cohort, further pointing to the critical role of the IL8-CXCR2 axis in AML/MDS. Functionally, CXCR2 inhibition by knockdown and pharmacologic approaches led to a significant reduction in proliferation in several leukemic cell lines and primary MDS/AML samples via induction of G0/G1 cell cycle arrest. Importantly, inhibition of CXCR2 selectively inhibited immature hematopoietic stem cells from MDS/AML samples without an effect on healthy controls. CXCR2 knockdown also impaired leukemic growth in vivo. Together, these studies demonstrate that the IL8 receptor CXCR2 is an adverse prognostic factor in MDS/AML and is a potential therapeutic target against immature leukemic stem cell-enriched cell fractions in MDS and AML.


Oncotarget | 2015

ASXL1 mutation correction by CRISPR/Cas9 restores gene function in leukemia cells and increases survival in mouse xenografts

Simona Valletta; Hamid Dolatshad; Matthias Bartenstein; Bon Ham Yip; Erica Bello; Shanisha Gordon; Yiting Yu; J Shaw; Swagata Roy; L Scifo; Anna Schuh; Andrea Pellagatti; Tudor A. Fulga; Amit Verma; Jacqueline Boultwood

Recurrent somatic mutations of the epigenetic modifier and tumor suppressor ASXL1 are common in myeloid malignancies, including chronic myeloid leukemia (CML), and are associated with poor clinical outcome. CRISPR/Cas9 has recently emerged as a powerful and versatile genome editing tool for genome engineering in various species. We have used the CRISPR/Cas9 system to correct the ASXL1 homozygous nonsense mutation present in the CML cell line KBM5, which lacks ASXL1 protein expression. CRISPR/Cas9-mediated ASXL1 homozygous correction resulted in protein re-expression with restored normal function, including down-regulation of Polycomb repressive complex 2 target genes. Significantly reduced cell growth and increased myeloid differentiation were observed in ASXL1 mutation-corrected cells, providing new insights into the role of ASXL1 in human myeloid cell differentiation. Mice xenografted with mutation-corrected KBM5 cells showed significantly longer survival than uncorrected xenografts. These results show that the sole correction of a driver mutation in leukemia cells increases survival in vivo in mice. This study provides proof-of-concept for driver gene mutation correction via CRISPR/Cas9 technology in human leukemia cells and presents a strategy to illuminate the impact of oncogenic mutations on cellular function and survival.


Journal of Clinical Investigation | 2017

The U2AF1S34F mutation induces lineage-specific splicing alterations in myelodysplastic syndromes

Bon Ham Yip; Violetta Steeples; Emmanouela Repapi; Richard N. Armstrong; Miriam Llorian; Swagata Roy; J Shaw; Hamid Dolatshad; Stephen Taylor; Amit Verma; Matthias Bartenstein; Paresh Vyas; Nicholas C.P. Cross; Luca Malcovati; Mario Cazzola; Eva Hellström-Lindberg; Seishi Ogawa; Christopher W. J. Smith; Andrea Pellagatti; Jacqueline Boultwood

Mutations of the splicing factor–encoding gene U2AF1 are frequent in the myelodysplastic syndromes (MDS), a myeloid malignancy, and other cancers. Patients with MDS suffer from peripheral blood cytopenias, including anemia, and an increasing percentage of bone marrow myeloblasts. We studied the impact of the common U2AF1S34F mutation on cellular function and mRNA splicing in the main cell lineages affected in MDS. We demonstrated that U2AF1S34F expression in human hematopoietic progenitors impairs erythroid differentiation and skews granulomonocytic differentiation toward granulocytes. RNA sequencing of erythroid and granulomonocytic colonies revealed that U2AF1S34F induced a higher number of cassette exon splicing events in granulomonocytic cells than in erythroid cells. U2AF1S34F altered mRNA splicing of many transcripts that were expressed in both cell types in a lineage-specific manner. In hematopoietic progenitors, the introduction of isoform changes identified in the U2AF1S34F target genes H2AFY, encoding an H2A histone variant, and STRAP, encoding serine/threonine kinase receptor–associated protein, recapitulated phenotypes associated with U2AF1S34F expression in erythroid and granulomonocytic cells, suggesting a causal link. Furthermore, we showed that isoform modulation of H2AFY and STRAP rescues the erythroid differentiation defect in U2AF1S34F MDS cells, suggesting that splicing modulators could be used therapeutically. These data have critical implications for understanding MDS phenotypic heterogeneity and support the development of therapies targeting splicing abnormalities.


Cancer Research | 2016

Pexmetinib: A Novel Dual Inhibitor of Tie2 and p38 MAPK with Efficacy in Preclinical Models of Myelodysplastic Syndromes and Acute Myeloid Leukemia.

Lohith S. Bachegowda; Kerry Morrone; Shannon L. Winski; Ioannis Mantzaris; Matthias Bartenstein; Nandini Ramachandra; Orsi Giricz; Vineeth Sukrithan; George Nwankwo; Samira Shahnaz; Tushar D. Bhagat; Sanchari Bhattacharyya; Amer Assal; Aditi Shastri; Shanisha Gordon-Mitchell; Andrea Pellagatti; Jacqueline Boultwood; Carolina Schinke; Yiting Yu; Chandan Guha; James P. Rizzi; Jennifer Garrus; Suzy Brown; Lance Wollenberg; Grant Hogeland; Dale Wright; Mark Munson; Mareli Rodriguez; Stefan Gross; David Chantry

Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) suppress normal hematopoietic activity in part by enabling a pathogenic inflammatory milieu in the bone marrow. In this report, we show that elevation of angiopoietin-1 in myelodysplastic CD34(+) stem-like cells is associated with higher risk disease and reduced overall survival in MDS and AML patients. Increased angiopoietin-1 expression was associated with a transcriptomic signature similar to known MDS/AML stem-like cell profiles. In seeking a small-molecule inhibitor of this pathway, we discovered and validated pexmetinib (ARRY-614), an inhibitor of the angiopoietin-1 receptor Tie-2, which was also found to inhibit the proinflammatory kinase p38 MAPK (which is overactivated in MDS). Pexmetinib inhibited leukemic proliferation, prevented activation of downstream effector kinases, and abrogated the effects of TNFα on healthy hematopoietic stem cells. Notably, treatment of primary MDS specimens with this compound stimulated hematopoiesis. Our results provide preclinical proof of concept for pexmetinib as a Tie-2/p38 MAPK dual inhibitor applicable to the treatment of MDS/AML. Cancer Res; 76(16); 4841-9. ©2016 AACR.


Stem cell investigation | 2016

Bone marrow fibrosis in primary myelofibrosis: pathogenic mechanisms and the role of TGF-β

Archana Agarwal; Kerry Morrone; Matthias Bartenstein; Zhizhuang Joe Zhao; Amit Verma; Swati Goel

Primary myelofibrosis (PMF) is a Philadelphia chromosome negative myeloproliferative neoplasm (MPN) with adverse prognosis and is associated with bone marrow fibrosis and extramedullary hematopoiesis. Even though the discovery of the Janus kinase 2 (JAK2), thrombopoietin receptor (MPL) and calreticulin (CALR) mutations have brought new insights into the complex pathogenesis of MPNs, the etiology of fibrosis is not well understood. Furthermore, since JAK2 inhibitors do not lead to reversal of fibrosis further understanding of the biology of fibrotic process is needed for future therapeutic discovery. Transforming growth factor beta (TGF-β) is implicated as an important cytokine in pathogenesis of bone marrow fibrosis. Various mouse models have been developed and have established the role of TGF-β in the pathogenesis of fibrosis. Understanding the molecular alterations that lead to TGF-β mediated effects on bone marrow microenvironment can uncover newer therapeutic targets against myelofibrosis. Inhibition of the TGF-β pathway in conjunction with other therapies might prove useful in the reversal of bone marrow fibrosis in PMF.


Cancer Research | 2017

Epigenetically Aberrant Stroma in MDS Propagates Disease via Wnt/β-Catenin Activation

Tushar D. Bhagat; Si Chen; Matthias Bartenstein; A. Trevor Barlowe; Dagny Von Ahrens; Gaurav S. Choudhary; Patrick Tivnan; Elianna Amin; A. Mario Marcondes; Mathijs A. Sanders; Remco M. Hoogenboezem; Suman Kambhampati; Nandini Ramachandra; Iaonnis Mantzaris; Vineeth Sukrithan; Remi Laurence; Robert Lopez; Prafullla Bhagat; Orsi Giricz; Davendra Sohal; Amittha Wickrema; Cecilia Yeung; Kira Gritsman; Peter D. Aplan; Yiting Yu; Kith Pradhan; Jinghang Zhang; John M. Greally; Siddhartha Mukherjee; Andrea Pellagatti

The bone marrow microenvironment influences malignant hematopoiesis, but how it promotes leukemogenesis has not been elucidated. In addition, the role of the bone marrow stroma in regulating clinical responses to DNA methyltransferase inhibitors (DNMTi) is also poorly understood. In this study, we conducted a DNA methylome analysis of bone marrow-derived stromal cells from myelodysplastic syndrome (MDS) patients and observed widespread aberrant cytosine hypermethylation occurring preferentially outside CpG islands. Stroma derived from 5-azacytidine-treated patients lacked aberrant methylation and DNMTi treatment of primary MDS stroma enhanced its ability to support erythroid differentiation. An integrative expression analysis revealed that the WNT pathway antagonist FRZB was aberrantly hypermethylated and underexpressed in MDS stroma. This result was confirmed in an independent set of sorted, primary MDS-derived mesenchymal cells. We documented a WNT/β-catenin activation signature in CD34+ cells from advanced cases of MDS, where it associated with adverse prognosis. Constitutive activation of β-catenin in hematopoietic cells yielded lethal myeloid disease in a NUP98-HOXD13 mouse model of MDS, confirming its role in disease progression. Our results define novel epigenetic changes in the bone marrow microenvironment, which lead to β-catenin activation and disease progression of MDS. Cancer Res; 77(18); 4846-57. ©2017 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Reduced DOCK4 expression leads to erythroid dysplasia in myelodysplastic syndromes.

Sriram Sundaravel; Ryan Duggan; Tushar D. Bhagat; David Ebenezer; Hui Liu; Yiting Yu; Matthias Bartenstein; Madhu Unnikrishnan; Subhradip Karmakar; Ting Chun Liu; Ingrid Torregroza; Thomas Quenon; John Anastasi; Kathy L. McGraw; Andrea Pellagatti; Jacqueline Boultwood; Vijay Yajnik; Andrew S. Artz; Michelle M. Le Beau; Ulrich Steidl; Alan F. List; Todd Evans; Amit Verma; Amittha Wickrema

Significance Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Genes that are aberrantly expressed and/or mutated that lead to the dysplastic erythroid morphology seen in −7/del(7q) MDS have not been identified. In this study, we show that reduced expression of dedicator of cytokinesis 4 (DOCK4) causes dysplasia by disrupting the actin cytoskeleton in developing red blood cells. In addition, our identification of the molecular pathway that leads to morphological defects in this type of MDS provides potential therapeutic targets downstream of DOCK4 that can be exploited to reverse the dysplasia in the erythroid lineage. Furthermore, we developed a novel single-cell multispectral flow cytometry assay for evaluation of disrupted F-actin filaments, which can be used for potential early detection of dysplastic cells in MDS. Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in −7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.


JCI insight | 2017

Efficacy of ALK5 inhibition in myelofibrosis

Lanzhu Yue; Matthias Bartenstein; Wanke Zhao; Wanting Tina Ho; Ying Han; Cem Murdun; Adam W. Mailloux; Ling Zhang; Xuefeng Wang; Anjali Budhathoki; Kith Pradhan; Franck Rapaport; Huaquan Wang; Zonghong Shao; Xiubao Ren; Ulrich Steidl; Ross L. Levine; Zhizhuang Joe Zhao; Amit Verma; Pearlie K. Epling-Burnette

Myelofibrosis (MF) is a bone marrow disorder characterized by clonal myeloproliferation, aberrant cytokine production, extramedullary hematopoiesis, and bone marrow fibrosis. Although somatic mutations in JAK2, MPL, and CALR have been identified in the pathogenesis of these diseases, inhibitors of the Jak2 pathway have not demonstrated efficacy in ameliorating MF in patients. TGF-β family members are profibrotic cytokines and we observed significant TGF-β1 isoform overexpression in a large cohort of primary MF patient samples. Significant overexpression of TGF-β1 was also observed in murine clonal MPLW515L megakaryocytic cells. TGF-β1 stimulated the deposition of excessive collagen by mesenchymal stromal cells (MSCs) by activating the TGF-β receptor I kinase (ALK5)/Smad3 pathway. MSCs derived from MPLW515L mice demonstrated sustained overproduction of both collagen I and collagen III, effects that were abrogated by ALK5 inhibition in vitro and in vivo. Importantly, use of galunisertib, a clinically active ALK5 inhibitor, significantly improved MF in both MPLW515L and JAK2V617F mouse models. These data demonstrate the role of malignant hematopoietic stem cell (HSC)/TGF-β/MSC axis in the pathogenesis of MF, and provide a preclinical rationale for ALK5 blockade as a therapeutic strategy in MF.


Journal of Clinical Investigation | 2018

Antisense STAT3 inhibitor decreases viability of myelodysplastic and leukemic stem cells

Aditi Shastri; Gaurav S. Choudhary; Margarida Teixeira; Shanisha Gordon-Mitchell; Nandini Ramachandra; Lumie Bernard; Sanchari Bhattacharyya; Robert Lopez; Kith Pradhan; Orsolya Giricz; Goutham Ravipati; Li-Fan Wong; Sally Cole; Tushar D. Bhagat; Jonathan Feld; Yosman Dhar; Matthias Bartenstein; Victor Thiruthuvanathan; Amittha Wickrema; B. Hilda Ye; David A. Frank; Andrea Pellagatti; Jacqueline Boultwood; Tianyuan Zhou; Youngsoo Kim; A. Robert MacLeod; Pearlie K. Epling-Burnette; Minwei Ye; Patricia McCoon; Richard Woessner

Acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) are associated with disease-initiating stem cells that are not eliminated by conventional therapies. Transcriptomic analysis of stem and progenitor populations in MDS and AML demonstrated overexpression of STAT3 that was validated in an independent cohort. STAT3 overexpression was predictive of a shorter survival and worse clinical features in a large MDS cohort. High STAT3 expression signature in MDS CD34+ cells was similar to known preleukemic gene signatures. Functionally, STAT3 inhibition by a clinical, antisense oligonucleotide, AZD9150, led to reduced viability and increased apoptosis in leukemic cell lines. AZD9150 was rapidly incorporated by primary MDS/AML stem and progenitor cells and led to increased hematopoietic differentiation. STAT3 knockdown also impaired leukemic growth in vivo and led to decreased expression of MCL1 and other oncogenic genes in malignant cells. These studies demonstrate that STAT3 is an adverse prognostic factor in MDS/AML and provide a preclinical rationale for studies using AZD9150 in these diseases.

Collaboration


Dive into the Matthias Bartenstein's collaboration.

Top Co-Authors

Avatar

Amit Verma

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tushar D. Bhagat

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yiting Yu

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kith Pradhan

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Orsolya Giricz

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

John M. Greally

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ulrich Steidl

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nandini Ramachandra

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge