Network


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

Hotspot


Dive into the research topics where Franz X. Schaub is active.

Publication


Featured researches published by Franz X. Schaub.


Blood | 2010

Clonal analysis of TET2 and JAK2 mutations suggests that TET2 can be a late event in the progression of myeloproliferative neoplasms.

Franz X. Schaub; Renate Looser; Sai Li; Hui Hao-Shen; Thomas Lehmann; André Tichelli; Radek C. Skoda

Somatic mutations in TET2 occur in patients with myeloproliferative neoplasms and other hematologic malignancies. It has been suggested that TET2 is a tumor suppressor gene and mutations in TET2 precede the acquisition of JAK2-V617F. To examine the order of events, we performed colony assays and genotyped TET2 and JAK2 in individual colonies. In 4 of 8 myeloproliferative neoplasm patients, we found that some colonies with mutated TET2 carried wild-type JAK2, whereas others were JAK2-V617F positive, indicating that TET2 occurred before JAK2-V617F. One of these patients carried a germline TET2 mutation. However, in 2 other patients, we obtained data compatible with the opposite order of events, with JAK2 exon 12 mutation preceding TET2 mutation in one case. Finally, in 2 of 8 patients, the TET2 and JAK2-V617F mutations defined 2 separate clones. The lack of a strict temporal order of occurrence makes it unlikely that mutations in TET2 represent a predisposing event for acquiring mutations in JAK2.


Haematologica | 2008

JAK2V617F mutation status identifies subtypes of refractory anemia with ringed sideroblasts associated with marked thrombocytosis

Annette Schmitt-Graeff; Soon-Siong Teo; Manfred Olschewski; Franz X. Schaub; Sabine Haxelmans; Andreas Kirn; Petra Reinecke; Ulrich Germing; Radek C. Skoda

Refractory anemia with ringed sideroblasts and marked thrombocytosis is a condition with both myelodysplastic and myeloproliferative features. This study indicates that a considerable proportion of patients with this condition carry the unique V617F mutation of JAK2, and that the mutant allele burden increases over time. See related perspective on page 4. Background Refractory anemia with ringed sideroblasts and marked thrombocytosis (RARS-T) was recently shown to be a JAK2-V617F mutation-related disorder. To determine the frequency and the prognostic significance of this mutation, we retrospectively evaluated 23 patients with platelet counts more than 600 × 109/L, 15% ringed sideroblasts or more, and at least erythroid marrow dysplasia. Design and Methods An allele-specific polymerase chain reaction for JAK2-V617F was used to determine the allelic ratio of the mutated JAK2 allele in DNA samples extracted from bone marrow biopsies. Hematologic and survival data of the JAK2-V617F positive vs. the JAK2-V617F negative patients were statistically analyzed. Allele-specific polymerase chain reaction was also used to screen for MPL-W515 mutations. Results The JAK2-V617F mutation was present in 11 patients (48%) and was associated with significantly higher erythrocyte and white blood cell counts (p=0.009 and 0.011, respectively). In 6/11 RARS-T patients the allelic ratio of JAK2-V617F was above 50%, indicating the presence of cells homozygous for the mutation. In two of these patients a transition from JAK2-V617F heterozygosity to homozygosity was documented and was accompanied by rising platelet counts in sequential samples. The MPL-W515L mutation was detected in one JAK2-V617F negative patient. The relative risk of death was found to be lower in the mutation-positive group than in the mutation-negative group. Conclusions RARS-T patients with JAK2-V617F have a more favorable prognosis than those without the JAK2 mutation. The prevalence of homozygous JAK2-V617F mutation in RARS-T suggests that this entity is biologically distinct from essential thrombocythemia.


Blood | 2009

Clonal analysis of deletions on chromosome 20q and JAK2-V617F in MPD suggests that del20q acts independently and is not one of the predisposing mutations for JAK2-V617F

Franz X. Schaub; Roland Jäger; Renate Looser; Hui Hao-Shen; Sylvie Hermouet; François Girodon; André Tichelli; Heinz Gisslinger; Robert Kralovics; Radek C. Skoda

We developed a real-time copy number polymerase chain reaction assay for deletions on chromosome 20q (del20q), screened peripheral blood granulocytes from 664 patients with myeloproliferative disorders, and identified 19 patients with del20q (2.9%), of which 14 (74%) were also positive for JAK2-V617F. To examine the temporal relationship between the occurrence of del20q and JAK2-V617F, we performed colony assays in methylcellulose, picked individual burst-forming units-erythroid (BFU-E) and colony-forming units-granulocyte (CFU-G) colonies, and genotyped each colony individually for del20q and JAK2-V617F. In 2 of 9 patients, we found that some colonies with del20q carried only wild-type JAK2, whereas other del20q colonies were JAK2-V617F positive, indicating that del20q occurred before the acquisition of JAK2-V617F. However, in colonies from 3 of 9 patients, we observed the opposite order of events. The lack of a strict temporal order of occurrence makes it doubtful that del20q represents a predisposing event for JAK2-V617F. In 2 patients with JAK2-V617F and 1 patient with MPL-W515L, microsatellite analysis revealed that del20q affected chromosomes of different parental origin and/or 9pLOH occurred at least twice. The fact that rare somatic events, such as del20q or 9pLOH, occurred more than once in subclones from the same patients suggests that the myeloproliferative disorder clone carries a predisposition to acquiring such genetic alterations.


Cell | 2012

Tristetraprolin Impairs Myc-Induced Lymphoma and Abolishes the Malignant State

Robert J. Rounbehler; Mohammad Fallahi; Chunying Yang; Meredith A. Steeves; Weimin Li; Joanne R. Doherty; Franz X. Schaub; Sandhya Sanduja; Dan A. Dixon; Perry J. Blackshear; John L. Cleveland

Myc oncoproteins directly regulate transcription by binding to target genes, yet this only explains a fraction of the genes affected by Myc. mRNA turnover is controlled via AU-binding proteins (AUBPs) that recognize AU-rich elements (AREs) found within many transcripts. Analyses of precancerous and malignant Myc-expressing B cells revealed that Myc regulates hundreds of ARE-containing (ARED) genes and select AUBPs. Notably, Myc directly suppresses transcription of Tristetraprolin (TTP/ZFP36), an mRNA-destabilizing AUBP, and this circuit is also operational during B lymphopoiesis and IL7 signaling. Importantly, TTP suppression is a hallmark of cancers with MYC involvement, and restoring TTP impairs Myc-induced lymphomagenesis and abolishes maintenance of the malignant state. Further, there is a selection for TTP loss in malignancy; thus, TTP functions as a tumor suppressor. Finally, Myc/TTP-directed control of select cancer-associated ARED genes is disabled during lymphomagenesis. Thus, Myc targets AUBPs to regulate ARED genes that control tumorigenesis.


Journal of Cell Biology | 2015

Hrr25/CK1δ-directed release of Ltv1 from pre-40S ribosomes is necessary for ribosome assembly and cell growth

Homa Ghalei; Franz X. Schaub; Joanne R. Doherty; Yoshihiko Noguchi; William R. Roush; John L. Cleveland; M. Elizabeth Stroupe; Katrin Karbstein

Cell growth relies on Hrr25/CK1δ-directed phosphorylation of Ltv1, which allows its release from nascent 40S ribosomal subunits and promotes subunit maturation.


Cancer Research | 2015

Fluorophore-NanoLuc BRET Reporters Enable Sensitive In Vivo Optical Imaging and Flow Cytometry for Monitoring Tumorigenesis

Franz X. Schaub; Md. Shamim Reza; Colin A. Flaveny; Weimin Li; Adele Musicant; Sany Hoxha; Min Guo; John L. Cleveland; Antonio L. Amelio

Fluorescent proteins are widely used to study molecular and cellular events, yet this traditionally relies on delivery of excitation light, which can trigger autofluorescence, photoxicity, and photobleaching, impairing their use in vivo. Accordingly, chemiluminescent light sources such as those generated by luciferases have emerged, as they do not require excitation light. However, current luciferase reporters lack the brightness needed to visualize events in deep tissues. We report the creation of chimeric eGFP-NanoLuc (GpNLuc) and LSSmOrange-NanoLuc (OgNLuc) fusion reporter proteins coined LumiFluors, which combine the benefits of eGFP or LSSmOrange fluorescent proteins with the bright, glow-type bioluminescent light generated by an enhanced small luciferase subunit (NanoLuc) of the deep-sea shrimp Oplophorus gracilirostris. The intramolecular bioluminescence resonance energy transfer that occurs between NanoLuc and the fused fluorophore generates the brightest bioluminescent signal known to date, including improved intensity, sensitivity, and durable spectral properties, thereby dramatically reducing image acquisition times and permitting highly sensitive in vivo imaging. Notably, the self-illuminating and bifunctional nature of these LumiFluor reporters enables greatly improved spatiotemporal monitoring of very small numbers of tumor cells via in vivo optical imaging and also allows the isolation and analyses of single cells by flow cytometry. Thus, LumiFluor reporters are inexpensive, robust, noninvasive tools that allow for markedly improved in vivo optical imaging of tumorigenic processes.


Blood | 2014

Myc-induced SUMOylation is a therapeutic vulnerability for B-cell lymphoma

Alexander Hoellein; Mohammad Fallahi; Stephanie Schoeffmann; Sabine Steidle; Franz X. Schaub; Martina Rudelius; Iina Laitinen; Lisa M. Nilsson; Andrei Goga; Christian Peschel; Jonas A. Nilsson; John L. Cleveland; Ulrich Keller

Myc oncogenic transcription factors (c-Myc, N-Myc, and L-Myc) coordinate the control of cell growth, division, and metabolism. In cancer, Myc overexpression is often associated with aggressive disease, which is in part due to the destruction of select targets by the ubiquitin-proteasome system (eg, SCF(Skp2)-directed destruction of the Cdk inhibitor p27(Kip1)). We reasoned that Myc would also regulate SUMOylation, a related means of posttranslational modification of proteins, and that this circuit would play essential roles in Myc-dependent tumorigenesis. Here, we report marked increases in the expression of genes that encode regulators and components of the SUMOylation machinery in mouse and human Myc-driven lymphomas, resulting in hyper-SUMOylation in these tumors. Further, inhibition of SUMOylation by genetic means disables Myc-induced proliferation, triggering G2/M cell-cycle arrest, polyploidy, and apoptosis. Using genetically defined cell models and conditional expression systems, this response was shown to be Myc specific. Finally, in vivo loss-of-function and pharmacologic studies demonstrated that inhibition of SUMOylation provokes rapid regression of Myc-driven lymphoma. Thus, targeting SUMOylation represents an attractive therapeutic option for lymphomas with MYC involvement.


Proceedings of the National Academy of Sciences of the United States of America | 2014

CRTC1/MAML2 gain-of-function interactions with MYC create a gene signature predictive of cancers with CREB–MYC involvement

Antonio L. Amelio; Mohammad Fallahi; Franz X. Schaub; Min Zhang; Mariam B. Lawani; Adam S. Alperstein; Mark R. Southern; Brandon Young; Lizi Wu; Maria Zajac-Kaye; Frederic J. Kaye; John L. Cleveland; Michael D. Conkright

Significance The prevailing dogma since the identification of the t (11, 19) translocation gene product as a fusion of the cAMP response element binding protein (CREB)-regulated transcriptional coactivator 1 (CRTC1) and the NOTCH coactivator mastermind-like 2 (MAML2) in malignant salivary gland tumors has been that aberrant activation of CREB and/or NOTCH transcription programs drives oncogenesis. However, combined expression of the parental coactivator molecules CRTC1 and MAML2 is not sufficient to induce transformation, suggesting an added level of complexity. Here we describe gain-of-function interactions between the CRTC1/MAML2 (C1/M2) coactivator fusion and myelocytomatosis oncogene (MYC) oncoproteins that are necessary for C1/M2-driven transformation. Our findings suggest that targeting the C1/M2–MYC interface represents an attractive strategy for the development of effective and safe anticancer therapeutics in tumors harboring the t (11, 19) translocation. Chimeric oncoproteins created by chromosomal translocations are among the most common genetic mutations associated with tumorigenesis. Malignant mucoepidermoid salivary gland tumors, as well as a growing number of solid epithelial-derived tumors, can arise from a recurrent t (11, 19)(q21;p13.1) translocation that generates an unusual chimeric cAMP response element binding protein (CREB)-regulated transcriptional coactivator 1 (CRTC1)/mastermind-like 2 (MAML2) (C1/M2) oncoprotein comprised of two transcriptional coactivators, the CRTC1 and the NOTCH/RBPJ coactivator MAML2. Accordingly, the C1/M2 oncoprotein induces aberrant expression of CREB and NOTCH target genes. Surprisingly, here we report a gain-of-function activity of the C1/M2 oncoprotein that directs its interactions with myelocytomatosis oncogene (MYC) proteins and the activation of MYC transcription targets, including those involved in cell growth and metabolism, survival, and tumorigenesis. These results were validated in human mucoepidermoid tumor cells that harbor the t (11, 19)(q21;p13.1) translocation and express the C1/M2 oncoprotein. Notably, the C1/M2–MYC interaction is necessary for C1/M2-driven cell transformation, and the C1/M2 transcriptional signature predicts other human malignancies having combined involvement of MYC and CREB. These findings suggest that such gain-of-function properties may also be manifest in other oncoprotein fusions found in human cancer and that agents targeting the C1/M2–MYC interface represent an attractive strategy for the development of effective and safe anticancer therapeutics in tumors harboring the t (11, 19) translocation.


Embo Molecular Medicine | 2014

Tipping the MYC-MIZ1 balance: targeting the HUWE1 ubiquitin ligase selectively blocks MYC-activated genes

Franz X. Schaub; John L. Cleveland

MYC family oncoproteins (MYC, N‐MYC and L‐MYC) function as basic helix‐loop‐helix‐leucine zipper (bHLH‐Zip) transcription factors that are activated (i.e., overexpressed) in well over half of all human malignancies (Boxer & Dang, ; Beroukhim et al, ). In this issue of EMBO Molecular Medicine, Eilers and colleagues (Peter et al, ) describe a novel approach to disable MYC, whereby inhibition of the ubiquitin ligase HUWE1 stabilizes MIZ1 and leads to the selective repression of MYC‐activated target genes.


Blood | 2011

Transition to homozygosity does not appear to provide a clonal advantage to hematopoietic progenitors carrying mutations in TET2

Franz X. Schaub; Thomas Lehmann; Renate Looser; Hui Hao-Shen; André Tichelli; Radek C. Skoda

To the editor: TET2 mutations are frequently found in hematologic malignancies including myeloproliferative neoplasms (MPNs).[1][1],[2][2] We previously reported on the clonal evolution of TET2 mutations in 8 JAK2 -V617F–positive MPN patients by genotyping single BFU-E colonies and found there is

Collaboration


Dive into the Franz X. Schaub's collaboration.

Top Co-Authors

Avatar

John L. Cleveland

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Carla Grandori

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Weimin Li

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Mohammad Fallahi

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Robert L. Diaz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonio L. Amelio

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Chunying Yang

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Michael Churchill

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Kemp

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge