Network


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

Hotspot


Dive into the research topics where Markus Müschen is active.

Publication


Featured researches published by Markus Müschen.


Nature | 2017

Metabolic gatekeeper function of B-lymphoid transcription factors

Lai N. Chan; Zhengshan Chen; Daniel Braas; Jae-Woong Lee; Gang Xiao; Huimin Geng; Kadriye Nehir Cosgun; Christian Hurtz; Seyedmehdi Shojaee; Valeria Cazzaniga; Hilde Schjerven; Thomas Ernst; Andreas Hochhaus; Steven M. Kornblau; Marina Konopleva; Miles A. Pufall; Giovanni Cazzaniga; Grace J. Liu; Thomas A. Milne; H. Phillip Koeffler; Theodora S. Ross; Isidro Sánchez-García; Arndt Borkhardt; Keith R. Yamamoto; Ross A. Dickins; Thomas G. Graeber; Markus Müschen

B-lymphoid transcription factors, such as PAX5 and IKZF1, are critical for early B-cell development, yet lesions of the genes encoding these transcription factors occur in over 80% of cases of pre-B-cell acute lymphoblastic leukaemia (ALL). The importance of these lesions in ALL has, until now, remained unclear. Here, by combining studies using chromatin immunoprecipitation with sequencing and RNA sequencing, we identify a novel B-lymphoid program for transcriptional repression of glucose and energy supply. Our metabolic analyses revealed that PAX5 and IKZF1 enforce a state of chronic energy deprivation, resulting in constitutive activation of the energy-stress sensor AMPK. Dominant-negative mutants of PAX5 and IKZF1, however, relieved this glucose and energy restriction. In a transgenic pre-B ALL mouse model, the heterozygous deletion of Pax5 increased glucose uptake and ATP levels by more than 25-fold. Reconstitution of PAX5 and IKZF1 in samples from patients with pre-B ALL restored a non-permissive state and induced energy crisis and cell death. A CRISPR/Cas9-based screen of PAX5 and IKZF1 transcriptional targets identified the products of NR3C1 (encoding the glucocorticoid receptor), TXNIP (encoding a glucose-feedback sensor) and CNR2 (encoding a cannabinoid receptor) as central effectors of B-lymphoid restriction of glucose and energy supply. Notably, transport-independent lipophilic methyl-conjugates of pyruvate and tricarboxylic acid cycle metabolites bypassed the gatekeeper function of PAX5 and IKZF1 and readily enabled leukaemic transformation. Conversely, pharmacological TXNIP and CNR2 agonists and a small-molecule AMPK inhibitor strongly synergized with glucocorticoids, identifying TXNIP, CNR2 and AMPK as potential therapeutic targets. Furthermore, our results provide a mechanistic explanation for the empirical finding that glucocorticoids are effective in the treatment of B-lymphoid but not myeloid malignancies. Thus, B-lymphoid transcription factors function as metabolic gatekeepers by limiting the amount of cellular ATP to levels that are insufficient for malignant transformation.


Journal of Experimental Medicine | 2017

Genetic analysis of Ikaros target genes and tumor suppressor function in BCR-ABL1+ pre–B ALL

Hilde Schjerven; Etapong F. Ayongaba; Ali Aghajanirefah; Jami McLaughlin; Donghui Cheng; Huimin Geng; Joseph R. Boyd; Linn M. Eggesbø; Ida Lindeman; Jessica L. Heath; Eugene Park; Owen N. Witte; Stephen T. Smale; Seth Frietze; Markus Müschen

Inactivation of the tumor suppressor gene encoding the transcriptional regulator Ikaros (IKZF1) is a hallmark of BCR-ABL1+ precursor B cell acute lymphoblastic leukemia (pre–B ALL). However, the mechanisms by which Ikaros functions as a tumor suppressor in pre–B ALL remain poorly understood. Here, we analyzed a mouse model of BCR-ABL1+ pre–B ALL together with a new model of inducible expression of wild-type Ikaros in IKZF1 mutant human BCR-ABL1+ pre–B ALL. We performed integrated genome-wide chromatin and expression analyses and identified Ikaros target genes in mouse and human BCR-ABL1+ pre–B ALL, revealing novel conserved gene pathways associated with Ikaros tumor suppressor function. Notably, genetic depletion of different Ikaros targets, including CTNND1 and the early hematopoietic cell surface marker CD34, resulted in reduced leukemic growth. Our results suggest that Ikaros mediates tumor suppressor function by enforcing proper developmental stage–specific expression of multiple genes through chromatin compaction at its target genes.


Nature Immunology | 2017

Antagonism of B cell enhancer networks by STAT5 drives leukemia and poor patient survival

Casey Katerndahl; Lynn M. Heltemes-Harris; Mark Willette; Christine Henzler; Seth Frietze; Rendong Yang; Hilde Schjerven; Kevin A. T. Silverstein; Laura B. Ramsey; Gregory Hubbard; Andrew D. Wells; Roland P. Kuiper; Blanca Scheijen; Frank N. van Leeuwen; Markus Müschen; Steven M. Kornblau; Michael A. Farrar

The transcription factor STAT5 has a critical role in B cell acute lymphoblastic leukemia (B-ALL). How STAT5 mediates this effect is unclear. Here we found that activation of STAT5 worked together with defects in signaling components of the precursor to the B cell antigen receptor (pre-BCR), including defects in BLNK, BTK, PKCβ, NF-κB1 and IKAROS, to initiate B-ALL. STAT5 antagonized the transcription factors NF-κB and IKAROS by opposing regulation of shared target genes. Super-enhancers showed enrichment for STAT5 binding and were associated with an opposing network of transcription factors, including PAX5, EBF1, PU.1, IRF4 and IKAROS. Patients with a high ratio of active STAT5 to NF-κB or IKAROS had more-aggressive disease. Our studies indicate that an imbalance of two opposing transcriptional programs drives B-ALL and suggest that restoring the balance of these pathways might inhibit B-ALL.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Circadian clock cryptochrome proteins regulate autoimmunity

Qi Cao; Xuan Zhao; Jingwen Bai; Sigal Gery; Haibo Sun; De-Chen Lin; Qi Chen; Zhengshan Chen; Lauren Mack; Henry Yang; Ruishu Deng; Xianping Shi; Ling Wa Chong; Han Cho; Jianjun Xie; Quan Zhen Li; Markus Müschen; Annette R. Atkins; Christopher Liddle; Ruth T. Yu; Serhan Alkan; Jonathan W. Said; Ye Zheng; Michael Downes; Ronald M. Evans; H. Phillip Koeffler

Significance Accumulated data show that circadian rhythms act as important regulators of specific immune functions, including activities of Toll-like receptor 9, macrophages, and Interleukin-17–producing CD4+ T helper (TH17) cells. In this study, we show that mice deficient of the circadian clock genes Cry1 and Cry2 unexpectedly display an autoimmune phenotype including high serum IgG concentrations, the presence of serum antinuclear antibodies, and precipitation of IgG, IgM, and complement 3 in glomeruli. Our results suggest that B cell development, the B cell-receptor–signaling pathway, and C1q expression are regulated by CRY proteins and that their dysregulation contributes to autoimmunity. The circadian system regulates numerous physiological processes including immune responses. Here, we show that mice deficient of the circadian clock genes Cry1 and Cry2 [Cry double knockout (DKO)] develop an autoimmune phenotype including high serum IgG concentrations, serum antinuclear antibodies, and precipitation of IgG, IgM, and complement 3 in glomeruli and massive infiltration of leukocytes into the lungs and kidneys. Flow cytometry of lymphoid organs revealed decreased pre-B cell numbers and a higher percentage of mature recirculating B cells in the bone marrow, as well as increased numbers of B2 B cells in the peritoneal cavity of Cry DKO mice. The B cell receptor (BCR) proximal signaling pathway plays a critical role in autoimmunity regulation. Activation of Cry DKO splenic B cells elicited markedly enhanced tyrosine phosphorylation of cellular proteins compared with cells from control mice, suggesting that overactivation of the BCR-signaling pathway may contribute to the autoimmunity phenotype in the Cry DKO mice. In addition, the expression of C1q, the deficiency of which contributes to the pathogenesis of systemic lupus erythematosus, was significantly down-regulated in Cry DKO B cells. Our results suggest that B cell development, the BCR-signaling pathway, and C1q expression are regulated by circadian clock CRY proteins and that their dysregulation through loss of CRY contributes to autoimmunity.


Cancer Research | 2018

Loss of Pax5 exploits Sca1-BCR-ABLp190 susceptibility to confer the metabolic shift essential for pB-ALL

Alberto Martín-Lorenzo; Franziska Auer; Lai N. Chan; Idoia García-Ramírez; Inés González-Herrero; Guillermo Rodríguez-Hernández; Christoph Bartenhagen; Martin Dugas; Michael Gombert; Sebastian Ginzel; Oscar Blanco; Alberto Orfao; Diego Alonso-López; Javier De Las Rivas; María Begoña García-Cenador; Francisco Javier García-Criado; Markus Müschen; Isidro Sánchez-García; Arndt Borkhardt; Carolina Vicente-Dueñas; Julia Hauer

Preleukemic clones carrying BCR-ABLp190 oncogenic lesions are found in neonatal cord blood, where the majority of preleukemic carriers do not convert into precursor B-cell acute lymphoblastic leukemia (pB-ALL). However, the critical question of how these preleukemic cells transform into pB-ALL remains undefined. Here, we model a BCR-ABLp190 preleukemic state and show that limiting BCR-ABLp190 expression to hematopoietic stem/progenitor cells (HS/PC) in mice (Sca1-BCR-ABLp190) causes pB-ALL at low penetrance, which resembles the human disease. pB-ALL blast cells were BCR-ABL-negative and transcriptionally similar to pro-B/pre-B cells, suggesting disease onset upon reduced Pax5 functionality. Consistent with this, double Sca1-BCR-ABLp190+Pax5+/- mice developed pB-ALL with shorter latencies, 90% incidence, and accumulation of genomic alterations in the remaining wild-type Pax5 allele. Mechanistically, the Pax5-deficient leukemic pro-B cells exhibited a metabolic switch toward increased glucose utilization and energy metabolism. Transcriptome analysis revealed that metabolic genes (IDH1, G6PC3, GAPDH, PGK1, MYC, ENO1, ACO1) were upregulated in Pax5-deficient leukemic cells, and a similar metabolic signature could be observed in human leukemia. Our studies unveil the first in vivo evidence that the combination between Sca1-BCR-ABLp190 and metabolic reprogramming imposed by reduced Pax5 expression is sufficient for pB-ALL development. These findings might help to prevent conversion of BCR-ABLp190 preleukemic cells.Significance: Loss of Pax5 drives metabolic reprogramming, which together with Sca1-restricted BCR-ABL expression enables leukemic transformation. Cancer Res; 78(10); 2669-79. ©2018 AACR.


Experimental Hematology | 2017

B-cell identity as a metabolic barrier against malignant transformation

Lai N. Chan; Markus Müschen

B-lineage and myeloid leukemia cells are often transformed by the same oncogenes, but have different biological and clinical characteristics. Although B-lineage acute lymphoblastic leukemia (B-ALL) cells are characterized by a state of chronic energy deficit, myeloid leukemia cells show abundant energy reserve. Interestingly, fasting has been demonstrated to inhibit selectively the development of B-ALL but not myeloid leukemia, further suggesting that lineage identity may be linked to divergent metabolic states in hematopoietic malignancies. The B-lymphoid transcription factors IKZF1, EBF1, and PAX5 are essential for early B-cell development and commitment to B-cell identity. However, in >80% of human pre-B-ALL cases, the leukemic clones harbor genetic lesions of these transcription factors. The significance of these defects has only recently been investigated. Here, we discuss the unexpected function of a B-lymphoid transcriptional program as a metabolic barrier against malignant transformation of B-cell precursor cells. The metabolic gatekeeper function of B-lymphoid transcription factors may force silent preleukemic clones carrying potentially oncogenic lesions to remain in a latent state. In addition, this program sets the threshold for responses to glucocorticoids in pre-B-ALL. Finally, the link between the tumor-suppressor and metabolic functions of B-lymphoid transcription factors is matched by observations in clinical trials: obesity and hyperglycemia are associated with poor clinical outcome in patients with pre-B-ALL.


Cell Reports | 2017

Lineage-Specific Genes Are Prominent DNA Damage Hotspots during Leukemic Transformation of B Cell Precursors

Bryant Boulianne; Mark E. Robinson; Philippa May; Leandro Castellano; Kevin Blighe; Jennifer Thomas; Alistair Reid; Markus Müschen; Jane F. Apperley; Justin Stebbing; Niklas Feldhahn

Summary In human leukemia, lineage-specific genes represent predominant targets of deletion, with lymphoid-specific genes frequently affected in lymphoid leukemia and myeloid-specific genes in myeloid leukemia. To investigate the basis of lineage-specific alterations, we analyzed global DNA damage in primary B cell precursors expressing leukemia-inducing oncogenes by ChIP-seq. We identified more than 1,000 sensitive regions, of which B lineage-specific genes constitute the most prominent targets. Identified hotspots at B lineage genes relate to DNA-DSBs, affect genes that harbor genomic lesions in human leukemia, and associate with ectopic deletion in successfully transformed cells. Furthermore, we show that most identified regions overlap with gene bodies of highly expressed genes and that induction of a myeloid lineage phenotype in transformed B cell precursors promotes de novo DNA damage at myeloid loci. Hence, we demonstrate that lineage-specific transcription predisposes lineage-specific genes in transformed B cell precursors to DNA damage, which is likely to promote the frequent alteration of lineage-specific genes in human leukemia.


Nature | 2018

Author Correction: Metabolic gatekeeper function of B-lymphoid transcription factors

Lai N. Chan; Zhengshan Chen; Daniel Braas; Jae-Woong Lee; Gang Xiao; Huimin Geng; Kadriye Nehir Cosgun; Christian Hurtz; Seyedmehdi Shojaee; Valeria Cazzaniga; Hilde Schjerven; Thomas Ernst; Andreas Hochhaus; Steven M. Kornblau; Marina Konopleva; Miles A. Pufall; Giovanni Cazzaniga; Grace J. Liu; Thomas A. Milne; H. Phillip Koeffler; Theodora S. Ross; Isidro Sánchez-García; Arndt Borkhardt; Keith R. Yamamoto; Ross A. Dickins; Thomas G. Graeber; Markus Müschen

Author(s): Chan, LN; Chen, Z; Braas, D; Lee, J-W; Xiao, G; Geng, H; Cosgun, KN; Hurtz, C; Shojaee, S; Cazzaniga, V; Schjerven, H; Ernst, T; Hochhaus, A; Kornblau, SM; Konopleva, M; Pufall, MA; Cazzaniga, G; Liu, GJ; Milne, TA; Koeffler, HP; Ross, TS; Sanchez-Garcia, I; Borkhardt, A; Yamamoto, KR; Dickins, RA; Graeber, TG; Muschen, M | Abstract: In Fig. 3c of this Letter, the the effects of CRISPR-Cas9-mediated deletion of NR3C1, TXNIP and CNR2 in patient-derived B-lineage leukaemia cells were shown. For curves depicting NR3C1 (left graph), data s for TXNIP (middle graph) were inadvertently plotted. This figure has been corrected online, and the original Fig. 3c is shown as Supplementary Information to this Amendment for transparency. The error does not affect the conclusions of the Letter. In addition, Source Data files have been added for the Figs. 1-4 and Extended Data Figs. 1-10 of the original Letter.In Fig. 3c of this Letter, the the effects of CRISPR–Cas9-mediated deletion of NR3C1, TXNIP and CNR2 in patient-derived B-lineage leukaemia cells were shown. Forxa0curves depictingxa0NR3C1 (leftxa0graph), data s for TXNIP (middlexa0graph)xa0were inadvertently plotted. This figure has been corrected online, and the original Fig. 3c is shown as Supplementary Information to this Amendmentxa0for transparency. The error does not affect the conclusions of the Letter. In addition, Source Data files have been added for the Figs. 1–4 and Extended Data Figs. 1–10 of the original Letter.


Journal of Hematology & Oncology | 2018

CAMKs support development of acute myeloid leukemia

Xunlei Kang; Changhao Cui; Chen Wang; Guojin Wu; Heyu Chen; Zhigang Lu; Xiaoli Chen; Li Wang; Jie Huang; Huimin Geng; Meng Zhao; Zhengshan Chen; Markus Müschen; Huan You Wang; Cheng Cheng Zhang

BackgroundWe recently identified the human leukocyte immunoglobulin-like receptor B2 (LILRB2) and its mouse ortholog-paired Ig-like receptor (PirB) as receptors for several angiopoietin-like proteins (Angptls). We also demonstrated that PirB is important for the development of acute myeloid leukemia (AML), but exactly how an inhibitory receptor such as PirB can support cancer development is intriguing.ResultsHere, we showed that the activation of Ca (2+)/calmodulin-dependent protein kinases (CAMKs) is coupled with PirB signaling in AML cells. High expression of CAMKs is associated with a poor overall survival probability in patients with AML. Knockdown of CAMKI or CAMKIV decreased human acute leukemia development in vitro and in vivo. Mouse AML cells that are defective in PirB signaling had decreased activation of CAMKs, and the forced expression of CAMK partially rescued the PirB-defective phenotype in the MLL-AF9 AML mouse model. The inhibition of CAMK kinase activity or deletion of CAMKIV significantly slowed AML development and decreased the AML stem cell activity. We also found that CAMKIV acts through the phosphorylation of one of its well-known target (CREB) in AML cells.ConclusionCAMKs are essential for the growth of human and mouse AML. The inhibition of CAMK signaling may become an effective strategy for treating leukemia.


Cell Reports | 2018

Simultaneous Targeting of PARP1 and RAD52 Triggers Dual Synthetic Lethality in BRCA-Deficient Tumor Cells

Katherine Sullivan-Reed; Elisabeth Bolton-Gillespie; Yashodhara Dasgupta; Samantha Langer; Micheal Siciliano; Margaret Nieborowska-Skorska; Kritika Hanamshet; Elizaveta A. Belyaeva; Andrea J. Bernhardy; Jaewong Lee; Morgan Moore; Huaqing Zhao; Peter Valent; Ksenia Matlawska-Wasowska; Markus Müschen; Smita Bhatia; Ravi Bhatia; Neil Johnson; Mariusz A. Wasik; Alexander V. Mazin; Tomasz Skorski

SUMMARY PARP inhibitors (PARPis) have been used to induce synthetic lethality in BRCA-deficient tumors in clinical trials with limited success. We hypothesized that RAD52-mediated DNA repair remains active in PARPi-treated BRCA-deficient tumor cells and that targeting RAD52 should enhance the synthetic lethal effect of PARPi. We show that RAD52 inhibitors (RAD52is) attenuated single-strand annealing (SSA) and residual homologous recombination (HR) in BRCA-deficient cells. Simultaneous targeting of PARP1 and RAD52 with inhibitors or dominant-negative mutants caused synergistic accumulation of DSBs and eradication of BRCA-deficient but not BRCA-proficient tumor cells. Remarkably, Parp1−/−; Rad52−/− mice are normal and display prolonged latency of BRCA1-deficient leukemia compared with Parp1−/− and Rad52−/− counterparts. Finally, PARPi+RAD52i exerted synergistic activity against BRCA1-deficient tumors in immunodeficient mice with minimal toxicity to normal cells and tissues. In conclusion, our data indicate that addition of RAD52i will improve therapeutic outcome of BRCA-deficient malignancies treated with PARPi.

Collaboration


Dive into the Markus Müschen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huimin Geng

University of California

View shared research outputs
Top Co-Authors

Avatar

Lai N. Chan

University of California

View shared research outputs
Top Co-Authors

Avatar

Zhengshan Chen

University of California

View shared research outputs
Top Co-Authors

Avatar

Steven M. Kornblau

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arndt Borkhardt

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

H. Phillip Koeffler

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge