FOXO transcription factors in hematopoietic tumors

ltr. Dr. Franziska Gehringer, Franz Ketzer, Anita Kick, Dr. Alexey Ushmorov

Research Interests

B cell lymphomas and B cell acute lymphoblastic leukemias (B-ALL) are malignancies that originate from B cells at different stages of their differentiation. B cell lymphomas and B-ALL belong to the most common hematological neoplasia. B-ALL has relatively good prognosis when treated with intensive chemotherapy according to current protocols (in 80-90% of children long-term remission can be achieved). Unfortunately, the cure rate after relapse is less than 40%. In addition, intensive chemotherapy often causes severe, sometimes life-threating acute and long-term side effects. In comparison with pediatric patients, the cure rates for adults suffering from B-ALL remain poor. Only 30-40 % of them achieve long-term remission. An impressive progress has also been achieved in the treatment of B cell lymphomas, but, as in case of B-ALLs, primary and acquired resistance to chemotherapy, and severe side effects of the intensive treatment protocols warrant searching for novel more efficient and less toxic therapeutic modalities. We are focusing on targeting B cell differentiation stage-specific proliferation and survival programs, which are maintained by most B cell malignancies.

Since 2010 we are investigating the role of FOXO transcription factors in the pathogenesis of B-cell neoplasia. FOXOs belong to the FOX O family of transcription factors, which share a highly conserved forkhead DNA -binding domain, but differ in the structure of transactivation domains which explains their partially non-redundant functions. The cellular functions of FOXOs include regulation of cell death, proliferation, protection from reactive oxygen species, DNA-damage response, and cellular metabolism (including lipogenesis and gluconeogenesis). Although members of the FOXO family are expressed in all tissues, there is an obvious quantitative difference in their tissue distribution. In particular, FOXO1 is highly expressed in B-cells. FOXO1 is essential for B cell survival and differentiation at early stages of B cell development, and for the germinal center reaction, which is responsible for affinity maturation of the B-cell receptor.

FOXO1 repression contributes to block of plasma cell differentiation in classical Hodgkin lymphoma

In 2012 (Xie L. et al., Blood, 2012) we paid attention on the low expression of FOXO1 in classical Hodgkin lymphoma in comparison with normal B cells and non-Hodgkin B cell lymphomas (cHL). cHL is a B-cell lymphoma which mainly lost its B-cell program. We have shown that overexpression of FOXO1 induced apoptosis in cHL cell lines and concluded that FOXO1 is a tumor suppressor in cHL. Latter we interrogated the molecular effects of FOXO1 overexpression in cHL with help of gene expression profiling. We identified the master regulator of plasma cell differentiation, PRDM1, as transcriptional target of FOXO1 which mediates its tumor suppressor function. Further, we demonstrated that the tumor-suppressor effect of PRDM1 depends on downregulation of the proto-oncogene MYC (Fig. 1). Moreover, we supposed that low FOXO1 expression is responsible for the characteristic “abortive plasma cell differentiation” phenotype of Hodgkin and Reed-Sternberg cells, the malignant component of cHL.

 

Fig. 1. FOXO1 repression contributes to the block of plasma cell differentiation in classical Hodgkin lymphoma (Vogel MJ et al. Blood 2014).

Fine-tuning of FOXO3A in cHL as a survival mechanism and a hallmark of abortive plasma cell differentiation

Continuing to investigate the role of FOXO1 in B cell lymphomas we paid attention on the reciprocal character of FOXO1 and FOXO3A mRNA expression in the process of mature B cell differentiation from germinal center dark zone (DZ) B cells to the plasma cells (PCs). The expression levels of FOXO1 decreased in the course of terminal differentiation from DZ B cell to PCs, whereas the FOXO1 mRNA levels increased. We noticed that FOXO3A mRNA levels in cHL were higher than in DZ B cells but lower than in PCs. We have further shown that this intermediate FOXO3A expression levels are essential for the survival of cHL, as either increase or decrease of the FOXO3A dosage was inappropriate (Fig. 2).

We concluded that cHL maintains FOXO3A/FOXO1 expression levels which reflect the differentiation status of the normal precursor cell of cHL, and that maintenance of the proliferation and survival program of this normal precursor is essential for the survival of cHL. In this way we postulated a Goldilocks principle for the FOXO effects in B cell lymphoma.

Fig. 2. Fine-tuning of FOXO3A in cHL as a survival mechanism and a hallmark of abortive plasma cell differentiation (Osswald CD, et al., 2018)

Tight regulation of FOXO1 is essential for maintenance of B-cell precursor acute lymphoblastic leukemia

Searching for support of our Goldilocks hypothesis we found that B-ALL, which is highly sensitive to FOXO-activating treatments (e.g. inhibitors of AKT or overexpression of FOXO1) maintain high FOXO1 expression in the nuclei, indicating a role of FOXO1 in B-ALL. We have shown that genetic depletion of FOXO1 induces growth arrest and apoptosis in B-ALL cell lines (Fig. 3). Most importantly, pharmacological inhibition of FOXO1 showed an anti-leukemia activity on several primary patient-derived pediatric ALL xenografts with effective leukemia reduction in the hematopoietic, lymphoid, and central nervous system organ compartments, ultimately leading to prolonged survival without leukemia reoccurrence in a preclinical in vivo model of BCP-ALL. These results suggested that repression of FOXO1 might be a feasible approach for the treatment of BCP-ALL.

Fig. 3. Tight regulation of FOXO1 is essential for maintenance of B-cell precursor acute lymphoblastic leukemia (Fan W. et al., Blood, 2018).

FOXO1 Confers Maintenance of the Dark Zone Proliferation and Survival Program and Can Be Pharmacologically Targeted in Burkitt Lymphoma

Re-analyzing our old data on FOXO1 expression in B cell non-Hodgkin lymphoma subtypes we identified high FOXO1 expression in nuclei of Burkitt lymphoma (BL). With help of loss-of-function experiments we identified an anti-proliferative effect of FOXO1 knockdown in BL cell lines, which was associated with the repression of the DZ B cell program including expression of MYB, CCND3, RAG2, BACH2, and CXCR4. In addition, the induction of signaling pathways of the light zone program like NF-κB and PI3K-AKT was observed. Using a rescue experiment we identified downregulation of the proto-oncogene MYB as a critical factor contributing to the antiproliferative effect of FOXO1 knockdown. In an attempt to estimate the feasibility of pharmacological FOXO1 repression, we found that the small molecular weight FOXO1 inhibitor AS1842856 induced cell death and growth arrest in BL cell lines at low concentrations. As in case of cHL and B-ALL, we found that overactivation of FOXO1 also induced growth inhibition in BL cell lines, indicating the importance of a tight regulation of the FOXO1 activity in BL (Fig. 4).

Fig. 4. FOXO1 confers maintenance of the dark zone proliferation and survival program and can be pharmacologically targeted in Burkitt lymphoma (Gehringer F., et al., Cancers, 2019).

Physiological levels of the PTEN-PI3K-AKT axis activity are required for maintenance of Burkitt lymphoma

Investigating the role of FOXO1 in the survival and proliferation program of BL, we also addressed the apparent paradox of the preferably nuclear FOXO1 expression and allegedly high activity of FOXO-inactivating PI3K-PDPK1-AKT pathway in BL. We found that the PI3K-PDPK1-AKT activity does not exceed physiological levels typical for germinal center B cells from which BL derives. Both overexpression of the constitutively active version of AKT (myrAKT) and knockdown of the natural PI3K-AKT signaling inhibitor PTEN resulted in FOXO1 inactivation and repressed the DZ proliferation and survival program and consequently the growth of BL (Fig. 5).

Fig. 5. Physiological levels of the PTEN-PI3K-AKT axis activity are required for maintenance of Burkitt lymphoma (Gehringer F., et al., Leukemia, 2020).

Our Main Findings

1. FOXO transcription factors and pathways modulating FOXO activity in B cell neoplasia cannot be considered as “tumor suppressors” and “oncogenes” in black-and-white terms.

2. FOXO activities are maintained in a ‘just right’ Goldilocks zone in certain B cell leukemia and lymphoma.

3. Selective pharmacological modulation of FOXO activities might be harnessed for the treatment of B cell leukemia and lymphoma.

CCND3 is essential for the maintenance of B-cell acute lymphoblastic leukemia

Previously we reported that inactivation of FOXO1 confers growth arrest and apoptosis in B-ALL, partially mediated by subsequent depletion of CCND3 (Wang F. et al., 2018). Given that previously the canonical MYC target CCND2 has been considered to play a major role in B-ALL proliferation, further investigation of the link between FOXO1 and CCND3 in B-ALL was warranted. In this study, we demonstrated that CCND3 is essential for the proliferation and survival of B-ALL, independent of the mutational background. Respectively, its expression at mRNA level exceeded that of CCND1 and CCND2. Furthermore, we identified FOXO1 as a CCND3-activating transcription factor in B-ALL. By comparing the effects of CCND3 depletion and CDK4/6 inhibition by palbociclib on B-ALL cells harboring different driver mutations, we found that the anti-apoptotic effect of CCND3 is independent of the kinase activity of the CCND3-CDK4/6 complex. Moreover, we found that CCND3 contributes to CDK8 transcription, which in part might explain the anti-apoptotic effect of CCND3. Finally, we found that increased CCND3 expression is associated with the development of resistance to palbociclib. We conclude that CCND3 plays an essential role in the maintenance of B-ALL, regardless of the underlying driver mutation. Moreover, downregulation of CCND3 expression might be superior to inhibition of CDK4/6 kinase activity in terms of B-ALL treatment.
 

Anti-apoptotic functions of CCND3 in B-ALL (Ketzer et al. 2022)

Collaborations

Peter Möller and Frank Leithäuser, Department of Pathology, University of Ulm, Ulm, Germany

Randy Gascoyne and Christian Steidl. Department of Pathology and Laboratory Medicine, Centre for Lymphoid Cancers and the Centre for Translational and Applied Genomics, Vancouver, Canada

Reuben Tooze, Section of Experimental Haematology, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK

Hassan Jumaa, Institut für Immunologie, Universitätsklinikum Ulm

Markus Müschen, Center of Molecular and Cellular Oncology, Yale School
of Medicine

Recent Publications

CCND3 is indispensable for the maintenance of B-cell acute lymphoblastic leukemia
Ketzer F, Abdelrasoul H, Vogel M, Marienfeld R, Müschen M, Jumaa H, Wirth T, Ushmorov A.
Oncogenesis. 2022 Jan 10;11(1):1.doi: 10.1038/s41389-021-00377-0.

Hyperstable SGK1 steps out of AKT’s shadow
Gehringer, F., & Ushmorov, A.
Blood.2021 Sep 16;138(11):917-918.doi: 10.1182/blood.2021012518

Physiological levels of the PTEN-PI3K-AKT axis activity are required for maintenance of Burkitt lymphoma.
Gehringer F, Weissinger SE, Möller P, Wirth T, Ushmorov A.
Leukemia. 2019 Nov 12. doi: 10.1038/s41375-019-0628-0.

FOXO1 Confers Maintenance of the Dark Zone Proliferation and Survival Program and Can Be Pharmacologically Targeted in Burkitt Lymphoma.
Gehringer F, Weissinger SE, Swier LJ, Möller P, Wirth T, Ushmorov A.
Cancers (Basel). 2019 Sep 25;11(10). pii: E1427. doi: 10.3390/cancers11101427.

FOXO in B-cell lymphopoiesis and B cell neoplasia.
Ushmorov A, Wirth T.
Semin Cancer Biol. 2018 Jun;50:132-141.

Tight regulation of FOXO1 is essential for maintenance of B-cell precursor acute lymphoblastic leukemia.
Wang F, Demir S, Gehringer F, Osswald CD, Seyfried F, Enzenmüller S, Eckhoff SM, Maier T, Holzmann K, Debatin KM, Wirth T, Meyer LH, Ushmorov A.
Blood. 2018 Apr 5. pii: blood-2017-10-813576. doi: 10.1182/blood-2017-10-813576. 

Fine-tuning of FOXO3A in cHL as a survival mechanism and a hallmark of abortive plasma cell differentiation.
Osswald CD, Xie L, Guan H, Herrmann F, Pick SM, Vogel MJ, Gehringer F, Chan FC, Steidl C, Wirth T, Ushmorov A.
Blood. 2018 Apr 5;131(14):1556-1567. doi: 10.1182/blood-2017-07-795278.

Activation of oncogenic pathways in classical Hodgkin lymphoma by decitabine: A rationale for combination with small molecular weight inhibitors.
Swerev TM, Wirth T, Ushmorov A.
Int J Oncol. 2017 Feb;50(2)

FOXO in B-cell lymphopoiesis and B cell neoplasia.
Ushmorov A, Wirth T.
Semin Cancer Biol. 2017 Jul 31. 

Contact

  • Fon: +49 (0)731/500-23271
    Fax: +49 (0)731/500-22892
  • Office
  •  
  • Postal Address:
  • University of Ulm
  • Institute of Physiological Chemistry
    Albert-Einstein-Allee 11
  • D-89081 Ulm

  • Suppliers Address:
  • University of Ulm
  • Institute of Physiological Chemistry
  • N27
    Meyerhofstrasse
    D-89081 Ulm