Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
EMBO J ; 41(7): e108397, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35156727

ABSTRACT

While PAX5 is an important tumor suppressor gene in B-cell acute lymphoblastic leukemia (B-ALL), it is also involved in oncogenic translocations coding for diverse PAX5 fusion proteins. PAX5-JAK2 encodes a protein consisting of the PAX5 DNA-binding region fused to the constitutively active JAK2 kinase domain. Here, we studied the oncogenic function of the PAX5-JAK2 fusion protein in a mouse model expressing it from the endogenous Pax5 locus, resulting in inactivation of one of the two Pax5 alleles. Pax5Jak2/+ mice rapidly developed an aggressive B-ALL in the absence of another cooperating exogenous gene mutation. The DNA-binding function and kinase activity of Pax5-Jak2 as well as IL-7 signaling contributed to leukemia development. Interestingly, all Pax5Jak2/+ tumors lost the remaining wild-type Pax5 allele, allowing efficient DNA-binding of Pax5-Jak2. While we could not find evidence for a nuclear role of Pax5-Jak2 as an epigenetic regulator, high levels of active phosphorylated STAT5 and increased expression of STAT5 target genes were seen in Pax5Jak2/+ B-ALL tumors, implying that nuclear Pax5-Jak2 phosphorylates STAT5. Together, these data reveal Pax5-Jak2 as an important nuclear driver of leukemogenesis by maintaining phosphorylated STAT5 levels in the nucleus.


Subject(s)
Janus Kinase 2 , Leukemia, B-Cell , PAX5 Transcription Factor , STAT5 Transcription Factor , Animals , Janus Kinase 2/genetics , Leukemia, B-Cell/genetics , Mice , Mutation , PAX5 Transcription Factor/genetics , STAT5 Transcription Factor/genetics , Translocation, Genetic
2.
EMBO J ; 36(6): 718-735, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28219927

ABSTRACT

PAX5 is a tumor suppressor in B-ALL, while the role of PAX5 fusion proteins in B-ALL development is largely unknown. Here, we studied the function of PAX5-ETV6 and PAX5-FOXP1 in mice expressing these proteins from the Pax5 locus. Both proteins arrested B-lymphopoiesis at the pro-B to pre-B-cell transition and, contrary to their proposed dominant-negative role, did not interfere with the expression of most regulated Pax5 target genes. Pax5-Etv6, but not Pax5-Foxp1, cooperated with loss of the Cdkna2a/b tumor suppressors in promoting B-ALL development. Regulated Pax5-Etv6 target genes identified in these B-ALLs encode proteins implicated in pre-B-cell receptor (BCR) signaling and migration/adhesion, which could contribute to the proliferation, survival, and tissue infiltration of leukemic B cells. Together with similar observations made in human PAX5-ETV6+ B-ALLs, these data identified PAX5-ETV6 as a potent oncoprotein that drives B-cell leukemia development.


Subject(s)
Oncogene Proteins/metabolism , PAX5 Transcription Factor/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-ets/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Oncogene Proteins/genetics , PAX5 Transcription Factor/genetics , Proto-Oncogene Proteins c-ets/genetics , Recombinant Fusion Proteins/genetics , Repressor Proteins/genetics , ETS Translocation Variant 6 Protein
3.
RNA ; 19(9): 1238-52, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23882114

ABSTRACT

Dicer is a key player in microRNA (miRNA) and RNA interference (RNAi) pathways, processing miRNA precursors and double-stranded RNA into ∼21-nt-long products ultimately triggering sequence-dependent gene silencing. Although processing of substrates in vertebrate cells occurs in the cytoplasm, there is growing evidence suggesting Dicer is also present and functional in the nucleus. To address this possibility, we searched for a nuclear localization signal (NLS) in human Dicer and identified its C-terminal double-stranded RNA binding domain (dsRBD) as harboring NLS activity. We show that the dsRBD-NLS can mediate nuclear import of a reporter protein via interaction with importins ß, 7, and 8. In the context of full-length Dicer, the dsRBD-NLS is masked. However, duplication of the dsRBD localizes the full-length protein to the nucleus. Furthermore, deletion of the N-terminal helicase domain results in partial accumulation of Dicer in the nucleus upon leptomycin B treatment, indicating that CRM1 contributes to nuclear export of Dicer. Finally, we demonstrate that human Dicer has the ability to shuttle between the nucleus and the cytoplasm. We conclude that Dicer is a shuttling protein whose steady-state localization is cytoplasmic.


Subject(s)
DEAD-box RNA Helicases/chemistry , Nuclear Localization Signals/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , Ribonuclease III/chemistry , Active Transport, Cell Nucleus , Binding Sites , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , DEAD-box RNA Helicases/metabolism , Humans , Nuclear Localization Signals/chemistry , Protein Transport , Ribonuclease III/metabolism , Transfection
4.
Nat Cancer ; 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103541

ABSTRACT

Combination approaches are needed to strengthen and extend the clinical response to KRASG12C inhibitors (KRASG12Ci). Here, we assessed the antitumor responses of KRASG12C mutant lung and colorectal cancer models to combination treatment with a SOS1 inhibitor (SOS1i), BI-3406, plus the KRASG12C inhibitor, adagrasib. We found that responses to BI-3406 plus adagrasib were stronger than to adagrasib alone, comparable to adagrasib with SHP2 (SHP2i) or EGFR inhibitors and correlated with stronger suppression of RAS-MAPK signaling. BI-3406 plus adagrasib treatment also delayed the emergence of acquired resistance and elicited antitumor responses from adagrasib-resistant models. Resistance to KRASG12Ci seemed to be driven by upregulation of MRAS activity, which both SOS1i and SHP2i were found to potently inhibit. Knockdown of SHOC2, a MRAS complex partner, partially restored response to KRASG12Ci treatment. These results suggest KRASG12C plus SOS1i to be a promising strategy for treating both KRASG12Ci naive and relapsed KRASG12C-mutant tumors.

5.
J Biol Chem ; 287(5): 3156-64, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22167198

ABSTRACT

The highly conserved DYNLL1 (LC8) protein was originally discovered as a light chain of the dynein motor complex, but is increasingly emerging as a sequence-specific regulator of protein dimerization with hundreds of targets and wide-ranging cellular functions. Despite its important roles, DYNLL1's own regulation remains poorly understood. Here we identify ASCIZ (ATMIN/ZNF822), an essential Zn(2+) finger protein with dual roles in the DNA base damage response and as a developmental transcription factor, as a conserved regulator of Dynll1 gene expression. DYNLL1 levels are reduced by ∼10-fold in the absence of ASCIZ in human, mouse and chicken cells. ASCIZ binds directly to the Dynll1 promoter and regulates its activity in a Zn(2+) finger-dependent manner. DYNLL1 protein in turn interacts with ten binding sites in the ASCIZ transcription activation domain, and high DYNLL1 levels inhibit the transcriptional activity of ASCIZ. In addition, DYNLL1 was also required for DNA damage-induced ASCIZ focus formation. The dual ability of ASCIZ to activate Dynll1 gene expression and to sense free DYNLL1 protein levels enables a simple dynamic feedback loop to adjust DYNLL1 levels to cellular needs. The ASCIZ-DYNLL1 feedback loop represents a novel mechanism for auto-regulation of gene expression, where the gene product directly inhibits the transcriptional activator while bound at its own promoter.


Subject(s)
Carrier Proteins/metabolism , Cytoplasmic Dyneins/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/physiology , Zinc/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Cell Line , Chickens , Cytoplasmic Dyneins/genetics , Humans , Mice , Nuclear Proteins/genetics , Transcription Factors , Transcription, Genetic/physiology , Zinc Fingers
6.
PLoS Genet ; 6(10): e1001170, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20975950

ABSTRACT

Zn²(+)-finger proteins comprise one of the largest protein superfamilies with diverse biological functions. The ATM substrate Chk2-interacting Zn²(+)-finger protein (ASCIZ; also known as ATMIN and ZNF822) was originally linked to functions in the DNA base damage response and has also been proposed to be an essential cofactor of the ATM kinase. Here we show that absence of ASCIZ leads to p53-independent late-embryonic lethality in mice. Asciz-deficient primary fibroblasts exhibit increased sensitivity to DNA base damaging agents MMS and H2O2, but Asciz deletion knock-down does not affect ATM levels and activation in mouse, chicken, or human cells. Unexpectedly, Asciz-deficient embryos also exhibit severe respiratory tract defects with complete pulmonary agenesis and severe tracheal atresia. Nkx2.1-expressing respiratory precursors are still specified in the absence of ASCIZ, but fail to segregate properly within the ventral foregut, and as a consequence lung buds never form and separation of the trachea from the oesophagus stalls early. Comparison of phenotypes suggests that ASCIZ functions between Wnt2-2b/ß-catenin and FGF10/FGF-receptor 2b signaling pathways in the mesodermal/endodermal crosstalk regulating early respiratory development. We also find that ASCIZ can activate expression of reporter genes via its SQ/TQ-cluster domain in vitro, suggesting that it may exert its developmental functions as a transcription factor. Altogether, the data indicate that, in addition to its role in the DNA base damage response, ASCIZ has separate developmental functions as an essential regulator of respiratory organogenesis.


Subject(s)
Carrier Proteins/physiology , DNA Repair/physiology , Lung/embryology , Nuclear Proteins/physiology , Organogenesis/physiology , Animals , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Cellular Senescence , DNA Damage , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Humans , Hydrogen Peroxide/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidants/pharmacology , Time Factors , Trachea/embryology , Transcription Factors , Ultraviolet Rays
7.
bioRxiv ; 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36747713

ABSTRACT

Efforts to improve the anti-tumor response to KRASG12C targeted therapy have benefited from leveraging combination approaches. Here, we compare the anti-tumor response induced by the SOS1-KRAS interaction inhibitor, BI-3406, combined with a KRASG12C inhibitor (KRASG12Ci) to those induced by KRASG12Ci alone or combined with SHP2 or EGFR inhibitors. In lung cancer and colorectal cancer (CRC) models, BI-3406 plus KRASG12Ci induces an anti-tumor response stronger than that observed with KRASG12Ci alone and comparable to those by the other combinations. This enhanced anti-tumor response is associated with a stronger and extended suppression of RAS-MAPK signaling. Importantly, BI-3406 plus KRASG12Ci treatment delays the emergence of acquired adagrasib resistance in both CRC and lung cancer models and is associated with re-establishment of anti-proliferative activity in KRASG12Ci-resistant CRC models. Our findings position KRASG12C plus SOS1 inhibition therapy as a promising strategy for treating both KRASG12C-mutated tumors as well as for addressing acquired resistance to KRASG12Ci.

8.
Cell Rep ; 14(6): 1488-1499, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26832406

ABSTRACT

How MYC promotes the development of cancer remains to be fully understood. Here, we report that the Zn(2+)-finger transcription factor ASCIZ (ATMIN, ZNF822) synergizes with MYC to activate the expression of dynein light chain (DYNLL1, LC8) in the murine Eµ-Myc model of lymphoma. Deletion of Asciz or Dynll1 prevented the abnormal expansion of pre-B cells in pre-cancerous Eµ-Myc mice and potentiated the pro-apoptotic activity of MYC in pre-leukemic immature B cells. Constitutive loss of Asciz or Dynll1 delayed lymphoma development in Eµ-Myc mice, and induced deletion of Asciz in established lymphomas extended the survival of tumor-bearing mice. We propose that ASCIZ-dependent upregulation of DYNLL1 levels is essential for the development and expansion of MYC-driven lymphomas by enabling the survival of pre-neoplastic and malignant cells.


Subject(s)
Dyneins/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Precursor Cells, B-Lymphoid/pathology , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Animals , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Cytoplasmic Dyneins , Disease Models, Animal , Dyneins/deficiency , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Mice , Precursor Cells, B-Lymphoid/immunology , Proto-Oncogene Proteins c-myc/immunology , Signal Transduction , Survival Analysis , Transcription Factors/deficiency
9.
J Exp Med ; 209(9): 1629-39, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22891272

ABSTRACT

Developing B lymphocytes expressing defective or autoreactive pre-B or B cell receptors (BCRs) are eliminated by programmed cell death, but how the balance between death and survival signals is regulated to prevent immunodeficiency and autoimmunity remains incompletely understood. In this study, we show that absence of the essential ATM (ataxia telangiectasia mutated) substrate Chk2-interacting Zn(2+)-finger protein (ASCIZ; also known as ATMIN/ZNF822), a protein with dual functions in the DNA damage response and as a transcription factor, leads to progressive cell loss from the pre-B stage onwards and severely diminished splenic B cell numbers in mice. This lymphopenia cannot be suppressed by deletion of p53 or complementation with a prearranged BCR, indicating that it is not caused by impaired DNA damage responses or defective V(D)J recombination. Instead, ASCIZ-deficient B cell precursors contain highly reduced levels of DYNLL1 (dynein light chain 1; LC8), a recently identified transcriptional target of ASCIZ, and normal B cell development can be restored by ectopic Dynll1 expression. Remarkably, the B cell lymphopenia in the absence of ASCIZ can also be fully suppressed by deletion of the proapoptotic DYNLL1 target Bim. Our findings demonstrate a key role for ASCIZ in regulating the survival of developing B cells by activating DYNLL1 expression, which may then modulate Bim-dependent apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , B-Lymphocytes/physiology , Carrier Proteins/metabolism , Dyneins/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , B-Lymphocytes/pathology , Bcl-2-Like Protein 11 , Carrier Proteins/genetics , Cytoplasmic Dyneins , DNA Damage , Dyneins/genetics , Gene Expression Regulation , Lymphopenia/genetics , Lymphopenia/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors , V(D)J Recombination
10.
Cell Cycle ; 10(8): 1222-4, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21415597

ABSTRACT

The ATM substrate Chk2-interacting Zn(2+)-finger protein (ASCIZ, also known as ATMIN and ZNF822) has previously been reported to be important for the repair of methylating and oxidative DNA damage, and it has also been proposed to regulate the stability and DNA damage-independent activation of the ATM kinase. While the role of the protein in the regulation of ATM remains controversial, two recent ASCIZ mouse knockout papers confirm its role in the DNA base damage response, including oxidative stress resistance in vivo. Similar to other DNA base damage repair proteins, ASCIZ is essential for embryonic development, with lethality of Asciz-null embryos around day E16.5 post conception. Unexpectedly, absence of ASCIZ also leads to severe organ development defects, most notably, complete absence of lungs similar to mutants in Wnt2-2b/ß-catenin and FGF10/FGFR2b signalling pathways. Together with evidence that ASCIZ can activate transcription in vitro, the phenotype indicates that ASCIZ has dual functions as an efficiency factor for DNA base damage repair as well as a key transcriptional regulator of early lung development.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Organogenesis/genetics , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , DNA Damage , DNA Methylation , DNA-Binding Proteins/genetics , Female , Gene Expression , Genetic Association Studies , Lung/embryology , Lung/metabolism , Mice , Mice, Knockout , Nuclear Proteins/genetics , Phenotype , Pregnancy , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Transcription Factors , Transcription, Genetic , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL