Network


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

Hotspot


Dive into the research topics where Jozef Madzo is active.

Publication


Featured researches published by Jozef Madzo.


Cancer Cell | 2011

Tet2 Loss Leads to Increased Hematopoietic Stem Cell Self-Renewal and Myeloid Transformation

Kelly Moran-Crusio; Linsey Reavie; Alan Shih; Omar Abdel-Wahab; Delphine Ndiaye-Lobry; Camille Lobry; Maria E. Figueroa; Aparna Vasanthakumar; Jay Patel; Xinyang Zhao; Fabiana Perna; Suveg Pandey; Jozef Madzo; Chun-Xiao Song; Qing Dai; Chuan He; Sherif Ibrahim; Miloslav Beran; Jiri Zavadil; Stephen D. Nimer; Ari Melnick; Lucy A. Godley; Iannis Aifantis; Ross L. Levine

Somatic loss-of-function mutations in the ten-eleven translocation 2 (TET2) gene occur in a significant proportion of patients with myeloid malignancies. Although there are extensive genetic data implicating TET2 mutations in myeloid transformation, the consequences of Tet2 loss in hematopoietic development have not been delineated. We report here an animal model of conditional Tet2 loss in the hematopoietic compartment that leads to increased stem cell self-renewal in vivo as assessed by competitive transplant assays. Tet2 loss leads to a progressive enlargement of the hematopoietic stem cell compartment and eventual myeloproliferation in vivo, including splenomegaly, monocytosis, and extramedullary hematopoiesis. In addition, Tet2(+/-) mice also displayed increased stem cell self-renewal and extramedullary hematopoiesis, suggesting that Tet2 haploinsufficiency contributes to hematopoietic transformation in vivo.


Cell Reports | 2014

TET1-Mediated Hydroxymethylation Facilitates Hypoxic Gene Induction in Neuroblastoma

Christopher J. Mariani; Aparna Vasanthakumar; Jozef Madzo; Ali Yesilkanal; Tushar D. Bhagat; Yiting Yu; Sanchari Bhattacharyya; Roland H. Wenger; Susan L. Cohn; Jayasri Nanduri; Amit Verma; Nanduri R. Prabhakar; Lucy A. Godley

SUMMARY The ten-eleven-translocation 5-methylcytosine dioxygenase (TET) family of enzymes catalyzes the conversion of 5-methylcytosine (5-mC) to 5-hydroxyme-thylcytosine (5-hmC), a modified cytosine base that facilitates gene expression. Cells respond to hypoxia by inducing a transcriptional program regulated in part by oxygen-dependent dioxygenases that require Fe(II) and α-ketoglutarate. Given that the TET enzymes also require these cofactors, we hypothesized that the TETs regulate the hypoxia-induced transcriptional program. Here, we demonstrate that hypoxia increases global 5-hmC levels, with accumulation of 5-hmC density at canonical hypoxia response genes. A subset of 5-hmC gains colocalize with hypoxia response elements facilitating DNA demethylation and HIF binding. Hypoxia results in transcriptional activation of TET1, and full induction of hypoxia-responsive genes and global 5-hmC increases require TET1. Finally, we show that 5-hmC increases and TET1 upregulation in hypoxia are HIF-1 dependent. These findings establish TET1-mediated 5-hmC changes as an important epigenetic component of the hypoxic response.


Cell Reports | 2014

Hydroxymethylation at Gene Regulatory Regions Directs Stem/Early Progenitor Cell Commitment during Erythropoiesis

Jozef Madzo; Hui Liu; Alexis Rodriguez; Aparna Vasanthakumar; Sriram Sundaravel; Donne Bennett D. Caces; Timothy J. Looney; Li Zhang; Janet B. Lepore; Trisha Macrae; Robert Duszynski; Alan H. Shih; Chun-Xiao Song; Miao Yu; Yiting Yu; Robert L. Grossman; Brigitte Raumann; Amit Verma; Chuan He; Ross L. Levine; Don Lavelle; Bruce T. Lahn; Amittha Wickrema; Lucy A. Godley

Hematopoietic stem cell differentiation involves the silencing of self-renewal genes and induction of a specific transcriptional program. Identification of multiple covalent cytosine modifications raises the question of how these derivatized bases influence stem cell commitment. Using a replicative primary human hematopoietic stem/progenitor cell differentiation system, we demonstrate dynamic changes of 5-hydroxymethylcytosine (5-hmC) during stem cell commitment and differentiation to the erythroid lineage. Genomic loci that maintain or gain 5-hmC density throughout erythroid differentiation contain binding sites for erythroid transcription factors and several factors not previously recognized as erythroid-specific factors. The functional importance of 5-hmC was demonstrated by impaired erythroid differentiation, with augmentation of myeloid potential, and disrupted 5-hmC patterning in leukemia patient-derived CD34+ stem/early progenitor cells with TET methylcytosine dioxygenase 2 (TET2) mutations. Thus, chemical conjugation and affinity purification of 5-hmC-enriched sequences followed by sequencing serve as resources for deciphering functional implications for gene expression during stem cell commitment and differentiation along a particular lineage.


Cancer Research | 2015

TET2 mutations affect non-CpG island DNA methylation at enhancers and transcription factor binding sites in chronic myelomonocytic leukemia

Jumpei Yamazaki; Jaroslav Jelinek; Yue Lu; Matteo Cesaroni; Jozef Madzo; Frank Neumann; Rong He; Rodolphe Taby; Aparna Vasanthakumar; Trisha Macrae; Kelly R. Ostler; Hagop M. Kantarjian; Shoudan Liang; Marcos R. Estecio; Lucy A. Godley; Jean-Pierre Issa

TET2 enzymatically converts 5-methylcytosine to 5-hydroxymethylcytosine as well as other covalently modified cytosines and its mutations are common in myeloid leukemia. However, the exact mechanism and the extent to which TET2 mutations affect DNA methylation remain in question. Here, we report on DNA methylomes in TET2 wild-type (TET2-WT) and mutant (TET2-MT) cases of chronic myelomonocytic leukemia (CMML). We analyzed 85,134 CpG sites [28,114 sites in CpG islands (CGI) and 57,020 in non-CpG islands (NCGI)]. TET2 mutations do not explain genome-wide differences in DNA methylation in CMML, and we found few and inconsistent differences at CGIs between TET2-WT and TET2-MT cases. In contrast, we identified 409 (0.71%) TET2-specific differentially methylated CpGs (tet2-DMCs) in NCGIs, 86% of which were hypermethylated in TET2-MT cases, suggesting a strikingly different biology of the effects of TET2 mutations at CGIs and NCGIs. DNA methylation of tet2-DMCs at promoters and nonpromoters repressed gene expression. Tet2-DMCs showed significant enrichment at hematopoietic-specific enhancers marked by H3K4me1 and at binding sites for the transcription factor p300. Tet2-DMCs showed significantly lower 5-hydroxymethylcytosine in TET2-MT cases. We conclude that leukemia-associated TET2 mutations affect DNA methylation at NCGI regions containing hematopoietic-specific enhancers and transcription factor-binding sites.


Cancers | 2013

Alterations of 5-Hydroxymethylcytosine in Human Cancers

Christopher J. Mariani; Jozef Madzo; Erika L. Moen; Ali Yesilkanal; Lucy A. Godley

Prior to 2009, 5-methylcytosine (5-mC) was thought to be the only biologically significant cytosine modification in mammalian DNA. With the discovery of the TET enzymes, which convert 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC), however, intense interest has emerged in determining the biological function of 5-hmC. Here, we review the techniques used to study 5-hmC and evidence that alterations to 5-hmC physiology play a functional role in the molecular pathogenesis of human cancers.


Scientific Reports | 2015

TET-catalyzed 5-hydroxymethylcytosine regulates gene expression in differentiating colonocytes and colon cancer.

Christopher G. Chapman; Christopher J. Mariani; Feng Wu; Katherine Meckel; Fatma Butun; Alice Chuang; Jozef Madzo; Marc B. Bissonnette; John H. Kwon; Lucy A. Godley

The formation of differentiated cell types from pluripotent progenitors involves epigenetic regulation of gene expression. DNA hydroxymethylation results from the enzymatic oxidation of 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC) by the ten-eleven translocation (TET) 5-mC dioxygenase enzymes. Previous work has mapped changes in 5-mC during differentiation of intestinal stem cells. However, whether or not 5-hmC regulates colonocyte differentiation is unknown. Here we show that 5-hmC regulates gene expression during colonocyte differentiation and controls gene expression in human colon cancers. Genome-wide profiling of 5-hmC during in vitro colonic differentiation demonstrated that 5-hmC is gained at highly expressed and induced genes and is associated with intestinal transcription factor binding sites, including those for HNF4A and CDX2. TET1 induction occurred during differentiation, and TET1 knockdown altered gene expression and inhibited barrier formation of colonocytes. We find that the 5-hmC distribution in primary human colonocytes parallels the distribution found in differentiated cells in vitro, and that gene-specific 5-hmC changes in human colon cancers are directly correlated with changes in gene expression. Our results support a model in which 5-hmC regulates differentiation of adult human intestine and 5-hmC alterations contribute to the disrupted gene expression in colon cancer.


Leukemia & Lymphoma | 2010

Identification and molecular characterization of a novel 3′ mutation in RUNX1 in a family with familial platelet disorder

Jane E. Churpek; Jacqueline S. Garcia; Jozef Madzo; Sarah Jackson; Kenan Onel; Lucy A. Godley

Familial platelet disorder with propensity to acute myelogenous leukemia (FPD/AML; OMIM 601399) is a rare Mendelian disorder characterized by clinical bleeding due to platelet dysfunction, mild to ...


Cancer Research | 2017

Transcriptional Selectivity of Epigenetic Therapy in Cancer

Takahiro Sato; Matteo Cesaroni; Woonbok Chung; Shoghag Panjarian; Anthony Tran; Jozef Madzo; Yasuyuki Okamoto; Hanghang Zhang; Xiaowei Chen; Jaroslav Jelinek; Jean-Pierre Issa

A central challenge in the development of epigenetic cancer therapy is the ability to direct selectivity in modulating gene expression for disease-selective efficacy. To address this issue, we characterized by RNA-seq, DNA methylation, and ChIP-seq analyses the epigenetic response of a set of colon, breast, and leukemia cancer cell lines to small-molecule inhibitors against DNA methyltransferases (DAC), histone deacetylases (Depsi), histone demethylases (KDM1A inhibitor S2101), and histone methylases (EHMT2 inhibitor UNC0638 and EZH2 inhibitor GSK343). We also characterized the effects of DAC as combined with the other compounds. Averaged over the cancer cell models used, we found that DAC affected 8.6% of the transcriptome and that 95.4% of the genes affected were upregulated. DAC preferentially regulated genes that were silenced in cancer and that were methylated at their promoters. In contrast, Depsi affected the expression of 30.4% of the transcriptome but showed little selectivity for gene upregulation or silenced genes. S2101, UNC0638, and GSK343 affected only 2% of the transcriptome, with UNC0638 and GSK343 preferentially targeting genes marked with H3K9me2 or H3K27me3, respectively. When combined with histone methylase inhibitors, the extent of gene upregulation by DAC was extended while still maintaining selectivity for DNA-methylated genes and silenced genes. However, the genes upregulated by combination treatment exhibited limited overlap, indicating the possibility of targeting distinct sets of genes based on different epigenetic therapy combinations. Overall, our results demonstrated that DNA methyltransferase inhibitors preferentially target cancer-relevant genes and can be combined with inhibitors targeting histone methylation for synergistic effects while still maintaining selectivity. Cancer Res; 77(2); 470-81. ©2016 AACR.


Seminars in Hematology | 2013

Perturbations of 5-Hydroxymethylcytosine Patterning in Hematologic Malignancies

Jozef Madzo; Aparna Vasanthakumar; Lucy A. Godley

The recent identification of covalent cytosine modifications derived from the metabolism of 5-methylcytosine (5-mC) and catalyzed by the TET proteins has facilitated molecular insight into a new subclass of acute myeloid leukemias (AMLs). TET2-mutant AMLs have the predicted hypermethylation phenotype expected given the inability of the mutant TET2 protein to convert 5-mC to 5-hydroxymethylcytosine (5-hmC). In addition, IDH1/2 mutations confer a gain-of-function, allowing the enzymes to process α-ketoglutarate to 2-hydroxyglutarate, which inhibits the TET proteins and ultimately induces the same hypermethylation phenotype. New techniques are being developed rapidly that have the unprecedented capacity to distinguish among the various covalent cytosine modifications now known to exist. Soon, these methods will be harnessed to yield a new level of insight into AMLs with altered distribution of 5-hmC, information that may allow new diagnostic and therapeutic approaches for patients with this subtype of AML.


Blood | 2012

Identifying familial myelodysplastic/acute leukemia predisposition syndromes through hematopoietic stem cell transplantation donors with thrombocytopenia

Jane E. Churpek; Eric Nickels; Rafael Marquez; Katarzyna Rojek; Bohao Liu; Rachelle Lorenz; Janet B. Lepore; Jozef Madzo; Amittha Wickrema; Andrew S. Artz; Koen van Besien; Lucy A. Godley

To the editor: The familial aggregation of thrombocytopenia and hematopoietic malignancies is seen in several autosomal-dominant familial myelodysplastic syndrome/acute leukemia predisposition syndromes (MDS/AL PS).[1][1] We hypothesized that some thrombocytopenic HLA-matched related subjects who

Collaboration


Dive into the Jozef Madzo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge