Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cell, Elsevier BV, Vol. 173, No. 2 ( 2018-04), p. 470-484.e18
    Type of Medium: Online Resource
    ISSN: 0092-8674
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 187009-9
    detail.hit.zdb_id: 2001951-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Nature, Springer Science and Business Media LLC, Vol. 558, No. 7711 ( 2018-6), p. E5-E5
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Nature, Springer Science and Business Media LLC, Vol. 542, No. 7642 ( 2017-02-23), p. 479-483
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 368-368
    Abstract: Introduction Activation during normal immune responses and oncogenic transformation impose increased metabolic demands on B-cells and their ability to retain redox homeostasis. While the serine/threonine-protein phosphatase 2A (PP2A) was identified as tumor suppressor in multiple types of cancer, our genetic studies revealed an unexpected lineage-specific dependency on PP2A in a broad range of B-cell malignancies. Results PP2A regulates glycolysis rate and balances energy supply against anti-oxidant protection of B-cells through the pentose-phosphate pathway (PPP). In contrast to robust PPP-activity in other hematopoietic lineages, B-lymphoid transcription factors (PAX5, IKZF1) restrict PPP-activity by transcriptional repression of G6PD and other rate-limiting PPP enzymes. Constitutively low PPP-activity and limited capacity to balance redox fluctuations cause a unique dependency on PP2A in B-cell tumors. Pharmacological ablation of PP2A and PPP-activity identify this pathway as a novel lineage-specific therapeutic target in a broad range of B-cell malignancies. Highlights • Conditional deletion of Ppp2r1a, the central scaffold assembling the PP2A holoenzyme, induces acute cell death of early and mature B-cells but does not affect other hematopoietic lineages. • PP2A redirects glucose carbon utilization from glycolysis to the PPP to mitigate oxidative stress. • B-cell malignancies exhibit constitutively low PPP activity, owing to B-cell-specific transcriptional repression of G6PD and other rate-limiting PPP enzymes. • Transcriptional repression of PPP activity in B-cells represents the mechanistic basis for the unique dependency of B-cell malignancies on PP2A. • Small molecule inhibitors of PP2A and G6PD act synergistically and overcome conventional drug-resistance in B-cell tumors. Summary Genetic lesions of PP2A are frequent in solid tumors and myeloid leukemia, but not in B-cell malignancies. Unlike other types of cancer, our genetic studies revealed an essential role of PP2A in B-cell tumors. Thereby, PP2A redirects glucose carbon utilization from glycolysis to PPP to salvage oxidative stress. This unique vulnerability reflects constitutively low PPP activity in B-cells and transcriptional repression of G6PD and other key PPP enzymes by the B-cell transcription factors PAX5 and IKZF1. Reflecting B-cell-specific transcriptional repression of PPP activity, glucose carbon utilization in B-cells is heavily skewed in favor of glycolysis resulting in lack of PPP-dependent antioxidant protection. These findings reveal a novel gatekeeper function of the PPP in a broad range of B-cell malignancies that can be efficiently targeted by small molecule inhibition of PP2A and G6PD. Citation Format: Gang Xiao, Zhengshan Chen, Lai N. Chan, Daniel Braas, Thomas G. Graeber, Huimin Geng, Hassan Jumaa, Xiaoyan Jiang, Markus Müschen. B-lymphoid transcriptional repression of the pentose phosphate pathway reveals a unique therapeutic vulnerability of B cell malignancies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 368.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Nature Medicine, Springer Science and Business Media LLC, Vol. 19, No. 8 ( 2013-8), p. 1014-1022
    Type of Medium: Online Resource
    ISSN: 1078-8956 , 1546-170X
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2013
    detail.hit.zdb_id: 1484517-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 783-783
    Abstract: Background & Hypothesis: The Philadelphia chromosome encoding the oncogenic BCR-ABL1 (Ph+) tyrosine kinase represents a common transforming oncogene in both chronic myeloid leukemia (CML) and a subset of pre-B acute lymphoblastic leukemia (ALL). While both diseases are driven by the same oncogene, biological and clinical characteristics differ between CML and Ph+ ALL. Given that oncogenic signaling from potent tyrosine kinases like BCR-ABL1 imposes significant metabolic requirements on energy supply, biogenesis and ability to survive metabolic stress, we investigated whether the divergent characteristics of BCR-ABL1 CML and Ph+ ALL have a metabolic basis. Results: Interestingly, metabolic analyses demonstrated that patient-derived CML chronic phase (CML-CP) and Ph+ ALL cells have distinguishable glycolytic profiles. Consistent with a higher proliferative rate, Ph+ ALL cells rapidly exhausted the capacity of their glycolytic machinery when compared to CML-CP cells. Furthermore, when comparing their glycolytic reserve (i.e. spare glycolytic capacity needed to compensate for loss of mitochondrial ATP production), Ph+ ALL cells were shown to have a lower glycolytic reserve compared to CML-CP cells. These findings suggest that Ph+ ALL cells have less flexibility in adapting to metabolic stress. Searching for factors that mediate metabolic reprogramming to support rapid cellular proliferation and promote adaptation to metabolic stress in a lineage-specific manner, we identified the tumor suppressor LKB1 and its substrate AMPK which coordinate metabolism with cell growth. To elucidate the potential lineage-specific functions of LKB1, genetic mouse models for 4-hydroxytamoxifen (4-OHT)-inducible deletion of Lkb1 in BCR-ABL1-driven myeloid leukemia (CML-like) and pre-B ALL (Ph+ ALL-like) were developed. Cre-mediated deletion of Lkb1 in myeloid leukemia mediated proliferation and stimulated aerobic glycolysis, as reflected by increases in both glucose consumption and lactate production. These findings are in agreement with previous findings in solid tumors and are consistent with the role of LKB1 as a tumor suppressor. In striking contrast, deletion of Lkb1 in pre-B ALL cells induced apoptosis and cell cycle arrest. In vivo, Lkb1 deletion in pre-B ALL cells delayed leukemia initiation and prolonged overall survival of transplant recipient mice. Furthermore, bioenergetics measurements of glycolytic and mitochondrial functions in response to various conditions of metabolic stress revealed that loss of Lkb1 function in pre-B ALL resulted in impaired metabolic adaptation. Likewise, metabolite profiling revealed that deletion of Lkb1 in pre-B ALL cells resulted in altered levels of metabolites from glycolysis, the TCA cycle, the pentose phosphate pathway, nucleotide metabolism, currency metabolites, amino acid metabolism and fatty acid metabolism. Importantly, C/EBPα-mediated lineage reprogramming of pre-B ALL cells into the myeloid lineage alleviated their dependency on Lkb1. Similar to observations made with Cre-mediated deletion of Lkb1 in a model for BCR-ABL1 pre-B ALL, small molecule inhibition of AMPK using BML-275 (an ATP-competitive inhibitor) induced apoptosis in patient-derived pre-B ALL cells. Moreover, BML-275 had deleterious effects on metabolic adaptation in patient-derived pre-B ALL cells, but not in CML-CP cells. Finally, BML-275 synergized with pharmacological inhibition of PI3K/Akt signaling in inhibiting glycolytic function and eradicating patient-derived pre-B ALL cells. Combining BML-275 and a PI3K inhibitor (BKM120) exerted significantly more potent inhibitory effect on pre-B ALL progression than each agent alone, prolonging the overall survival of recipient mice. Conclusions: Taken together, our findings demonstrate that LKB1/AMPK signaling plays lineage-specific roles in BCR-ABL1-driven CML and pre-B ALL. While LKB1/AMPK is an established tumor suppressor pathway in multiple cell types, our findings reveal a unique and previously unrecognized vulnerability of pre-B ALL cells. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 1255-1255
    Abstract: Oncogenic lesions in hematopoietic progenitor cells give rise to B-cell or myeloid malignancies. While often transformed by the same oncogenes, B-cell and myeloid leukemias markedly differ in biological and clinical characteristics. Our metabolic analyses revealed that B-cell-unlike myeloid-leukemia cells are massively restricted in their glycolytic capacity. Low glycolytic reserves in B cells resulted in a state of chronic energy depletion and engaged the energy sensor LKB1-AMPK. Myeloid cells strongly activated glucose transport through insulin receptor (INSR)-AKT signaling and lacked activity of LKB1-AMPK, reflecting energy abundance. Conversely, B-cells lacked INSR-AKT signaling and were critically dependent on LKB1-AMPK-mediated glucose uptake. Cre-mediated deletion of Lkb1 caused acute glycolytic exhaustion and cell death in B-lineage but increased glycolysis, energy levels and proliferation in myeloid leukemia. C/EBPa-mediated conversion of B-cell into myeloid identity reversed the detrimental effects of Lkb1-deletion and restored glycolysis, energy levels and survival of B→myeloid reprogrammed cells. In 〉 80% of B-lineage leukemia cases, we found genetic lesions of transcription factors (e.g. deletion of PAX5, IKZF1, rearrangement of MLL) that caused a B→myeloid lineage shift. While previously of unknown functional significance, these lesions relieved B-cell-specific transcriptional repression of molecules that mediate glucose uptake and utilization (INSR, GLUT1, HK2, G6PD) and amplified glycolytic energy supply for transforming oncogenes. Likewise, glucocorticoid receptor (NR3C1)-mediated inhibition of glucose uptake and glycolysis was strictly dependent on a B-lymphoid transcriptional program. B→myeloid lineage conversion abolished NR3C1 expression and activity, which provides a mechanistic explanation for the empiric finding that glucocorticoids are highly active in the treatment of B-cell-but not myeloid malignancies. In conclusion, B-cell-specific restriction of glycolytic energy supply represents a previously unrecognized metabolic barrier against malignant transformation and reveals LKB1-AMPK as a novel target for the treatment of human B-lineage leukemia. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1124-1124
    Abstract: Oncogenic lesions in multi-potent progenitor cells often give rise to either B-cell or myeloid lineage leukemia. While transformed by the same oncogenes (e.g. BCR-ABL1, RAS), B-lineage and myeloid leukemias are distinct diseases. Given that oncogenic tyrosine kinase signaling (e.g. BCR-ABL1) imposes significant metabolic requirements on energy supply, biogenesis and metabolic fitness, we studied whether the divergent characteristics of myeloid and B-lineage leukemias have a metabolic basis. Metabolic analyses revealed that B-lineage acute lymphoblastic leukemia (Ph+ ALL) cells proliferate at maximum capacity of their glycolytic machinery. In contrast to myeloid leukemia (CML), B-lineage ALL cells lack metabolic adaptive fitness in response to metabolic fluctuations. C/EBPα-mediated reprogramming of B-lineage cells into the myeloid lineage induced glycolytic gene expression (Insr, Slc2a1, G6pdx, G6pd2, and Hk3). Frequent genetic lesions of transcription factors that determine B cell identity (IKZF1, PAX5, EBF1) partially mitigate B cell-instrinsic metabolic liability. Reconstitution of PAX5 expression in patient-derived B-lineage ALL cells reduced metabolic fitness by impacting glucose metabolism. Using genetic and metabolic experiments, we identified the metabolic liability observed in B-lineage ALL is in part dependent on the serine/threonine kinase LKB1. In agreement with previous studies, Cre-mediated deletion of Lkb1 induced proliferation in myeloid leukemia. Surprisingly, Lkb1 deletion led to apoptosis and decreased leukemogenic capacity in B-lineage leukemia. Consistent with the above observations, Arf, p53 and p27 levels were reduced in Lkb1-deficient myeloid leukemia cells, while Lkb1 deletion in B-lineage ALL cells up-regulated Arf, p53 and p27 levels. Enhanced glucose consumption and lactate production were observed in Lkb1-deficient myeloid leukemia cells. In contrast, loss of Lkb1 led to defective glycolytic and mitochondrial activity in B-lineage ALL. Lkb1 deletion in B-lineage ALL caused global accumulation of metabolites, suggesting that LKB1 is required for maintaining metabolic homeostasis. Moreover, loss of Lkb1 decreased protein levels of mitochondrial, anti-apoptotic BCL-2 family proteins, BCL-xL and MCL1, in B-lineage ALL. Reverse Phase Protein Array analyses revealed that LKB1 levels positively correlated with BCL-xL and MCL1 in patient-derived Ph+ ALL samples (n = 51) as well as other subtypes of B-lineage ALL (n = 183; MDACC, 1983-2007). Importantly, C/EBPα-mediated reprogramming of B-lineage ALL cells to the myeloid linage relieved dependency on LKB1. Taken together, we showed that transcriptional control of B cell identity causes unique metabolic liability. B-lineage ALL cells exhibit unique reliance on LKB1 for metabolic homeostasis and survival. Our findings revealed LKB1 as a potential therapeutic target in B-lineage ALL. Note: This abstract was not presented at the meeting. Citation Format: Lai N. Chan, Daniel Braas, Christian Hurtz, Seyedmehdi Shojaee, Huimin Geng, Valeria Cazzaniga, Carina Ng, Behzad Kharabi Masouleh, Yi Hua Qiu, Nianxiang Zhang, Kevin R. Coombes, Thomas Ernst, Giovanni Cazzaniga, Andreas Hochhaus, Steven Kornblau, Thomas Graeber. Transcriptional control of B cell identity restricts metabolic fitness in human leukemia. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1124. doi:10.1158/1538-7445.AM2015-1124
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages