Network


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

Hotspot


Dive into the research topics where Tihomira I. Todorova is active.

Publication


Featured researches published by Tihomira I. Todorova.


Blood | 2012

Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS

Laura Barreyro; Britta Will; Boris Bartholdy; Li Zhou; Tihomira I. Todorova; Robert F. Stanley; Susana Ben-Neriah; Cristina Montagna; Samir Parekh; Andrea Pellagatti; Jacqueline Boultwood; Elisabeth Paietta; Rhett P. Ketterling; Larry D. Cripe; Hugo F. Fernandez; Peter L. Greenberg; Martin S. Tallman; Christian Steidl; Constantine S. Mitsiades; Amit Verma; Ulrich Steidl

Cellular and interpatient heterogeneity and the involvement of different stem and progenitor compartments in leukemogenesis are challenges for the identification of common pathways contributing to the initiation and maintenance of acute myeloid leukemia (AML). Here we used a strategy of parallel transcriptional analysis of phenotypic long-term hematopoietic stem cells (HSCs), short-term HSCs, and granulocyte-monocyte progenitors from individuals with high-risk (-7/7q-) AML and compared them with the corresponding cell populations from healthy controls. This analysis revealed dysregulated expression of 11 genes, including IL-1 receptor accessory protein (IL1RAP), in all leukemic stem and progenitor cell compartments. IL1RAP protein was found to be overexpressed on the surface of HSCs of AML patients, and marked cells with the -7/7q- anomaly. IL1RAP was also overexpressed on HSCs of patients with normal karyotype AML and high-risk myelodysplastic syndrome, suggesting a pervasive role in different disease subtypes. High IL1RAP expression was independently associated with poor overall survival in 3 independent cohorts of AML patients (P = 2.2 × 10(-7)). Knockdown of IL1RAP decreased clonogenicity and increased cell death of AML cells. Our study identified genes dysregulated in stem and progenitor cells in -7/7q- AML, and suggests that IL1RAP may be a promising therapeutic and prognostic target in AML and high-risk myelodysplastic syndrome.


Nature Immunology | 2013

Satb1 regulates the self-renewal of hematopoietic stem cells by promoting quiescence and repressing differentiation commitment

Britta Will; Thomas O. Vogler; Boris Bartholdy; Francine E. Garrett-Bakelman; Jillian Mayer; Laura Barreyro; Ashley Pandolfi; Tihomira I. Todorova; Ujunwa C. Okoye-Okafor; Robert F. Stanley; Tushar D. Bhagat; Amit Verma; Maria E. Figueroa; Ari Melnick; Michael Roth; Ulrich Steidl

How hematopoietic stem cells (HSCs) coordinate the regulation of opposing cellular mechanisms such as self-renewal and differentiation commitment remains unclear. Here we identified the transcription factor and chromatin remodeler Satb1 as a critical regulator of HSC fate. HSCs lacking Satb1 had defective self-renewal, were less quiescent and showed accelerated lineage commitment, which resulted in progressive depletion of functional HSCs. The enhanced commitment was caused by less symmetric self-renewal and more symmetric differentiation divisions of Satb1-deficient HSCs. Satb1 simultaneously repressed sets of genes encoding molecules involved in HSC activation and cellular polarity, including Numb and Myc, which encode two key factors for the specification of stem-cell fate. Thus, Satb1 is a regulator that promotes HSC quiescence and represses lineage commitment.


Nature Medicine | 2015

Minimal PU.1 reduction induces a preleukemic state and promotes development of acute myeloid leukemia

Britta Will; Thomas O. Vogler; Swathi Rao Narayanagari; Boris Bartholdy; Tihomira I. Todorova; Mariana da Silva Ferreira; Jiahao Chen; Yiting Yu; Jillian Mayer; Laura Barreyro; Luis Carvajal; Daniela Ben Neriah; Michael Roth; Johanna van Oers; Sonja Schaetzlein; Christine McMahon; Winfried Edelmann; Amit Verma; Ulrich Steidl

Modest transcriptional changes caused by genetic or epigenetic mechanisms are frequent in human cancer. Although loss or near-complete loss of the hematopoietic transcription factor PU.1 induces acute myeloid leukemia (AML) in mice, a similar degree of PU.1 impairment is exceedingly rare in human AML; yet, moderate PU.1 inhibition is common in AML patients. We assessed functional consequences of modest reductions in PU.1 expression on leukemia development in mice harboring DNA lesions resembling those acquired during human stem cell aging. Heterozygous deletion of an enhancer of PU.1, which resulted in a 35% reduction of PU.1 expression, was sufficient to induce myeloid-biased preleukemic stem cells and their subsequent transformation to AML in a DNA mismatch repair–deficient background. AML progression was mediated by inhibition of expression of a PU.1-cooperating transcription factor, Irf8. Notably, we found marked molecular similarities between the disease in these mice and human myelodysplastic syndrome and AML. This study demonstrates that minimal reduction of a key lineage-specific transcription factor, which commonly occurs in human disease, is sufficient to initiate cancer development, and it provides mechanistic insight into the formation and progression of preleukemic stem cells in AML.


Journal of Clinical Investigation | 2014

HSC commitment–associated epigenetic signature is prognostic in acute myeloid leukemia

Boris Bartholdy; Maximilian Christopeit; Britta Will; Yongkai Mo; Laura Barreyro; Yiting Yu; Tushar D. Bhagat; Ujunwa C. Okoye-Okafor; Tihomira I. Todorova; John M. Greally; Ross L. Levine; Ari Melnick; Amit Verma; Ulrich Steidl

Acute myeloid leukemia (AML) is characterized by disruption of HSC and progenitor cell differentiation. Frequently, AML is associated with mutations in genes encoding epigenetic modifiers. We hypothesized that analysis of alterations in DNA methylation patterns during healthy HSC commitment and differentiation would yield epigenetic signatures that could be used to identify stage-specific prognostic subgroups of AML. We performed a nano HpaII-tiny-fragment-enrichment-by-ligation-mediated-PCR (nanoHELP) assay to compare genome-wide cytosine methylation profiles between highly purified human long-term HSC, short-term HSC, common myeloid progenitors, and megakaryocyte-erythrocyte progenitors. We observed that the most striking epigenetic changes occurred during the commitment of short-term HSC to common myeloid progenitors and these alterations were predominantly characterized by loss of methylation. We developed a metric of the HSC commitment–associated methylation pattern that proved to be highly prognostic of overall survival in 3 independent large AML patient cohorts, regardless of patient treatment and epigenetic mutations. Application of the epigenetic signature metric for AML prognosis was superior to evaluation of commitment-based gene expression signatures. Together, our data define a stem cell commitment–associated methylome that is independently prognostic of poorer overall survival in AML.


Journal of Clinical Investigation | 2017

Pharmacological inhibition of the transcription factor PU.1 in leukemia

Iléana Antony-Debré; Ananya Paul; Joana Leite; Kelly Mitchell; Hye Mi Kim; Luis Carvajal; Tihomira I. Todorova; Kenneth Huang; Arvind Kumar; Abdelbasset A. Farahat; Boris Bartholdy; Swathi Rao Narayanagari; Jiahao Chen; Alberto Ambesi-Impiombato; Adolfo A. Ferrando; Ioannis Mantzaris; Evripidis Gavathiotis; Amit Verma; Britta Will; David W. Boykin; W. David Wilson; Gregory M.K. Poon; Ulrich Steidl

The transcription factor PU.1 is often impaired in patients with acute myeloid leukemia (AML). Here, we used AML cells that already had low PU.1 levels and further inhibited PU.1 using either RNA interference or, to our knowledge, first-in-class small-molecule inhibitors of PU.1 that we developed specifically to allosterically interfere with PU.1-chromatin binding through interaction with the DNA minor groove that flanks PU.1-binding motifs. These small molecules of the heterocyclic diamidine family disrupted the interaction of PU.1 with target gene promoters and led to downregulation of canonical PU.1 transcriptional targets. shRNA or small-molecule inhibition of PU.1 in AML cells from either PU.1lo mutant mice or human patients with AML-inhibited cell growth and clonogenicity and induced apoptosis. In murine and human AML (xeno)transplantation models, treatment with our PU.1 inhibitors decreased tumor burden and resulted in increased survival. Thus, our study provides proof of concept that PU.1 inhibition has potential as a therapeutic strategy for the treatment of AML and for the development of small-molecule inhibitors of PU.1.


Science Translational Medicine | 2018

Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia

Luis Carvajal; Daniela Ben Neriah; Adrien Senecal; Lumie Benard; Victor Thiruthuvanathan; Tatyana Yatsenko; Swathi Rao Narayanagari; Justin C. Wheat; Tihomira I. Todorova; Kelly Mitchell; Charles Kenworthy; Vincent Guerlavais; D. Allen Annis; Boris Bartholdy; Britta Will; Jesus Anampa; Ioannis Mantzaris; Manuel Aivado; Robert H. Singer; Robert A. Coleman; Amit Verma; Ulrich Steidl

Dual inhibition of MDMX and MDM2 by an α-helical p53-stapled peptide (ALRN-6924) results in robust antitumor activity in acute myeloid leukemia. A new staple of leukemia treatment? As suggested by their name, tumor suppressor genes prevent tumorigenesis, and their expression or activity is often lost in cancer cells. One of the best known tumor suppressors is p53, which is inactivated in a variety of cancer types, often through up-regulation of its endogenous suppressors. Despite numerous attempts to reactivate p53 by a variety of approaches, none have successfully advanced beyond clinical trials thus far. Now, Carvajal et al. applied yet another tactic to restore p53 activity by using a stapled peptide to inactivate both of its endogenous inhibitors, for situations where the tumor suppressor is inactive but not completely lost. The authors demonstrated the effectiveness of this approach in human acute myeloid leukemia using in vitro and in vivo models, along with preliminary testing in a patient with leukemia. The tumor suppressor p53 is often inactivated via its interaction with endogenous inhibitors mouse double minute 4 homolog (MDM4 or MDMX) or mouse double minute 2 homolog (MDM2), which are frequently overexpressed in patients with acute myeloid leukemia (AML) and other cancers. Pharmacological disruption of both of these interactions has long been sought after as an attractive strategy to fully restore p53-dependent tumor suppressor activity in cancers with wild-type p53. Selective targeting of this pathway has thus far been limited to MDM2-only small-molecule inhibitors, which lack affinity for MDMX. We demonstrate that dual MDMX/MDM2 inhibition with a stapled α-helical peptide (ALRN-6924), which has recently entered phase I clinical testing, produces marked antileukemic effects. ALRN-6924 robustly activates p53-dependent transcription at the single-cell and single-molecule levels and exhibits biochemical and molecular biological on-target activity in leukemia cells in vitro and in vivo. Dual MDMX/MDM2 inhibition by ALRN-6924 inhibits cellular proliferation by inducing cell cycle arrest and apoptosis in cell lines and primary AML patient cells, including leukemic stem cell–enriched populations, and disrupts functional clonogenic and serial replating capacity. Furthermore, ALRN-6924 markedly improves survival in AML xenograft models. Our study provides mechanistic insight to support further testing of ALRN-6924 as a therapeutic approach in AML and other cancers with wild-type p53.


Molecular Cancer Therapeutics | 2013

Abstract C225: IL1RAP as functionally relevant target for stem-cell directed therapy in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS).

Laura Barreyro; Kelly Mitchell; Britta Will; Boris Bartholdy; Li Zhou; Tihomira I. Todorova; Robert F. Stanley; Susana Ben-Neriah; Cristina Montagna; Samir Parekh; Andrea Pellagatti; Jacqueline Boultwood; Elisabeth Paietta; Rhett P. Ketterling; Larry D. Cripe; Hugo F. Fernandez; Peter L. Greenberg; Jacob M. Rowe; Martin S. Tallman; Christian Steidl; Constantine S. Mitsiades; Amit Verma; Ulrich Steidl

Despite the established use of poly-chemotherapy, relapse continues to be the most common cause of death in AML and MDS and cure rates remain below 20%. AML/MDS arise following the accumulation of stepwise genetic and epigenetic changes in hematopoietic stem and progenitor cells (HSPC). Utilizing a novel strategy of parallel transcriptional analysis of sorted HSPC populations in distinct subtypes of AML, we compared the gene expression in AML HSPC with identical compartments from age-matched healthy controls and identified Interleukin 1 receptor accessory protein (IL1RAP) as one of the most significantly upregulated genes in HSPC in all examined subtypes of AML. Fluorescence in situ hybridization of sorted IL1RAP+ and IL1RAP- cells from patients with monosomy 7 AML (-7) indicated that the aberrant clone was restricted to IL1RAP+ cells, demonstrating that IL1RAP overexpression is a distinguishing feature of the -7 clone. Multivariate analysis of a large cohort of patients with normal karyotype AML showed that patients with high IL1RAP levels had inferior overall survival than patients with lower IL1RAP levels, suggesting an independent prognostic value for this molecule in AML. IL1RAP expression levels in MDS were found elevated on stem cells of patients with high risk disease, proposing a role of IL1RAP in higher risk MDS and progression to AML. Downregulation of IL1RAP expression by lentivirally expressed shRNAs decreased clonogenicity in cell lines and AML/MDS primary patient samples, induced apoptosis of AML cells, and reduced proliferation of AML cells and infiltration of hematopoietic organs in vivo. IL1RAP is a transmembrane protein required for signaling through several receptors of the IL1 family. Downregulation of IL1RAP expression in AML cells led to phosphorylation changes in several kinases and their substrates suggesting participation of IL1RAP in multiple signaling pathways and highlighting its potential as therapeutic target. We investigated whether inhibition of IL1RAP with pharmacological compounds is feasible and effective. Antibody-mediated inhibition of IL1RAP led to inhibition of AML cell growth in vitro. In addition, we designed peptides to interfere with IL1RAP-receptor interactions which lead to inhibition of AML cell growth. Both types of agents are being further tested and optimized. In summary, our study reveals IL1RAP as aberrantly expressed on HSPC of AML and high-risk MDS patients. Inhibition of IL1RAP is feasible and functionally effective, and thus has the potential to lead to novel therapies specifically directed at such stem cells. Beyond IL1RAP, our study provides a map of dysregulated transcripts in HSPC from patients with AML, which may offer further opportunities for therapeutic intervention. The strategy of comparative analysis of sorted stem and progenitor cells in cancer versus healthy controls may be applicable to other type of cancers with a suspected stem cell origin, and instrumental for the identification of targets for stem cell-directed therapy. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C225. Citation Format: Laura Barreyro, Kelly Mitchell, Britta Will, Boris Bartholdy, Li Zhou, Tihomira Todorova, Robert Stanley, Susana Ben-Neriah, Cristina Montagna, Samir Parekh, Andrea Pellagatti, Jacqueline Boultwood, Elisabeth Paietta, Rhett Ketterling, Larry Cripe, Hugo Fernandez, Peter Greenberg, Jacob Rowe, Martin Tallman, Christian Steidl, Constantine Mitsiades, Amit Verma, Ulrich Steidl. IL1RAP as functionally relevant target for stem-cell directed therapy in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr C225.


Journal of Experimental Medicine | 2018

IL1RAP potentiates multiple oncogenic signaling pathways in AML

Kelly Mitchell; Laura Barreyro; Tihomira I. Todorova; Samuel Taylor; Iléana Antony-Debré; Swathi-Rao Narayanagari; Luis Carvajal; Joana Leite; Zubair Piperdi; Gopichand Pendurti; Ioannis Mantzaris; Elisabeth Paietta; Amit Verma; Kira Gritsman; Ulrich Steidl

The surface molecule interleukin-1 receptor accessory protein (IL1RAP) is consistently overexpressed across multiple genetic subtypes of acute myeloid leukemia (AML) and other myeloid malignancies, including at the stem cell level, and is emerging as a novel therapeutic target. However, the cell-intrinsic functions of IL1RAP in AML cells are largely unknown. Here, we show that targeting of IL1RAP via RNA interference, genetic deletion, or antibodies inhibits AML pathogenesis in vitro and in vivo, without perturbing healthy hematopoietic function or viability. Furthermore, we found that the role of IL1RAP is not restricted to the IL-1 receptor pathway, but that IL1RAP physically interacts with and mediates signaling and pro-proliferative effects through FLT3 and c-KIT, two receptor tyrosine kinases with known key roles in AML pathogenesis. Our study provides a new mechanistic basis for the efficacy of IL1RAP targeting in AML and reveals a novel role for this protein in the pathogenesis of the disease.


Science Translational Medicine | 2018

Thrombopoietin receptor–independent stimulation of hematopoietic stem cells by eltrombopag

Yun-Ruei Kao; Jiahao Chen; Swathi-Rao Narayanagari; Tihomira I. Todorova; Maria M. Aivalioti; Mariana da Silva Ferreira; Pedro Ramos; Celine Pallaud; Ioannis Mantzaris; Aditi Shastri; James B. Bussel; Amit Verma; Ulrich Steidl; Britta Will

Eltrombopag stimulates multilineage hematopoiesis through intracellular iron chelation. Ironing out bone marrow failure Chronic thrombocytopenia can be associated with a variety of conditions, such as bone narrow failure syndromes and immune disorders. It increases the risk of severe bleeding, and therapies such as platelet transfusion or recombinant thrombopoietin are associated with a variety of complications. In contrast, the small-molecule eltrombopag, a thrombopoietin receptor antagonist, has been very effective in treating thrombocytopenia in patients with bone marrow failure. Kao et al. demonstrated that, in addition to targeting the thrombopoietin receptor, eltrombopag also chelates iron and that this chelating action enables it to improve the function of bone marrow stem cells. The authors also determined the mechanism linking iron chelation to hematopoietic stem cell maintenance, which may help facilitate the development of future treatments. Eltrombopag (EP), a small-molecule thrombopoietin receptor (TPO-R) agonist and potent intracellular iron chelator, has shown remarkable efficacy in stimulating sustained multilineage hematopoiesis in patients with bone marrow failure syndromes, suggesting an effect at the most immature hematopoietic stem and multipotent progenitor level. Although the functional and molecular effects of EP on megakaryopoiesis have been studied in the past, mechanistic insights into its effects on the earliest stages of hematopoiesis have been limited. We investigated the effects of EP treatment on hematopoietic stem cell (HSC) function using purified primary HSCs in separation-of-function mouse models, including a TPO-R–deficient strain, and stem cells isolated from patients undergoing TPO-R agonist treatment. Our mechanistic studies showed a stimulatory effect on stem cell self-renewal independently of TPO-R. Human and mouse HSCs responded to acute EP treatment with metabolic and gene expression alterations consistent with a reduction of intracellular labile iron pools that are essential for stem cell maintenance. Iron preloading prevented the stem cell stimulatory effects of EP. Moreover, comparative analysis of stem cells in the bone marrow of patients receiving EP showed a marked increase in the number of functional stem cells compared to patients undergoing therapy with romiplostim, another TPO-R agonist lacking an iron-chelating ability. Together, our study demonstrates that EP stimulates hematopoiesis at the stem cell level through iron chelation–mediated molecular reprogramming and indicates that labile iron pool–regulated pathways can modulate HSC function.


Journal of Experimental Medicine | 2017

A myeloid tumor suppressor role for NOL3

Robert F. Stanley; Richard T. Piszczatowski; Boris Bartholdy; Kelly Mitchell; Wendy M. McKimpson; Swathi Rao Narayanagari; Dagmar Walter; Tihomira I. Todorova; Cassandra M. Hirsch; Hideki Makishima; Britta Will; Christine McMahon; Kira Gritsman; Jaroslaw P. Maciejewski; Richard N. Kitsis; Ulrich Steidl

Despite the identification of several oncogenic driver mutations leading to constitutive JAK–STAT activation, the cellular and molecular biology of myeloproliferative neoplasms (MPN) remains incompletely understood. Recent discoveries have identified underlying disease-modifying molecular aberrations contributing to disease initiation and progression. Here, we report that deletion of Nol3 (Nucleolar protein 3) in mice leads to an MPN resembling primary myelofibrosis (PMF). Nol3−/− MPN mice harbor an expanded Thy1+LSK stem cell population exhibiting increased cell cycling and a myelomonocytic differentiation bias. Molecularly, this phenotype is mediated by Nol3−/−-induced JAK–STAT activation and downstream activation of cyclin-dependent kinase 6 (Cdk6) and Myc. Nol3−/− MPN Thy1+LSK cells share significant molecular similarities with primary CD34+ cells from PMF patients. NOL3 levels are decreased in CD34+ cells from PMF patients, and the NOL3 locus is deleted in a subset of patients with myeloid malignancies. Our results reveal a novel genetic PMF-like mouse model and identify a tumor suppressor role for NOL3 in the pathogenesis of myeloid malignancies.

Collaboration


Dive into the Tihomira I. Todorova's collaboration.

Top Co-Authors

Avatar

Ulrich Steidl

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Britta Will

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Boris Bartholdy

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Amit Verma

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Laura Barreyro

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Robert F. Stanley

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ujunwa C. Okoye-Okafor

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge