Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Blood Adv ; 5(7): 2027-2039, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33847741

ABSTRACT

CC-122 is a next-generation cereblon E3 ligase-modulating agent that has demonstrated promising clinical efficacy in patients with relapsed or refractory diffuse large B-cell lymphoma (R/R DLBCL). Mechanistically, CC-122 induces the degradation of IKZF1/3, leading to T-cell activation and robust cell-autonomous killing in DLBCL. We report a genome-wide CRISPR/Cas9 screening for CC-122 in a DLBCL cell line SU-DHL-4 with follow-up mechanistic characterization in 6 DLBCL cell lines to identify genes regulating the response to CC-122. Top-ranked CC-122 resistance genes encode, not only well-defined members or regulators of the CUL4/DDB1/RBX1/CRBN E3 ubiquitin ligase complex, but also key components of signaling and transcriptional networks that have not been shown to modulate the response to cereblon modulators. Ablation of CYLD, NFKBIA, TRAF2, or TRAF3 induces hyperactivation of the canonical and/or noncanonical NF-κB pathways and subsequently diminishes CC-122-induced apoptosis in 5 of 6 DLBCL cell lines. Depletion of KCTD5, the substrate adaptor of the CUL3/RBX1/KCTD5 ubiquitin ligase complex, promotes the stabilization of its cognate substrate, GNG5, resulting in CC-122 resistance in HT, SU-DHL-4, and WSU-DLCL2. Furthermore, knockout of AMBRA1 renders resistance to CC-122 in SU-DHL-4 and U-2932, whereas knockout of RFX7 leads to resistance specifically in SU-DHL-4. The ubiquitous and cell line-specific mechanisms of CC-122 resistance in DLBCL cell lines revealed in this work pinpoint genetic alternations that are potentially associated with clinical resistance in patients and facilitate the development of biomarker strategies for patient stratification, which may improve clinical outcomes of patients with R/R DLBCL.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Piperidones , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Potassium Channels , Quinazolinones , Ubiquitin-Protein Ligases
2.
Blood Cancer J ; 10(2): 16, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029705

ABSTRACT

Large-scale chromosomal translocations are frequent oncogenic drivers in acute myeloid leukemia (AML). These translocations often occur in critical transcriptional/epigenetic regulators and contribute to malignant cell growth through alteration of normal gene expression. Despite this knowledge, the specific gene expression alterations that contribute to the development of leukemia remain incompletely understood. Here, through characterization of transcriptional regulation by the RUNX1-ETO fusion protein, we have identified Ras-association domain family member 2 (RASSF2) as a critical gene that is aberrantly transcriptionally repressed in t(8;21)-associated AML. Re-expression of RASSF2 specifically inhibits t(8;21) AML development in multiple models. Through biochemical and functional studies, we demonstrate RASSF2-mediated functions to be dependent on interaction with Hippo kinases, MST1 and MST2, but independent of canonical Hippo pathway signaling. Using proximity-based biotin labeling we define the RASSF2-proximal proteome in leukemia cells and reveal association with Rac GTPase-related proteins, including an interaction with the guanine nucleotide exchange factor, DOCK2. Importantly, RASSF2 knockdown impairs Rac GTPase activation, and RASSF2 expression is broadly correlated with Rac-mediated signal transduction in AML patients. Together, these data reveal a previously unappreciated mechanistic link between RASSF2, Hippo kinases, and Rac activity with potentially broad functional consequences in leukemia.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/prevention & control , Oncogene Proteins, Fusion/metabolism , Translocation, Genetic , Tumor Suppressor Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Oncogene Proteins, Fusion/genetics , RNA, Long Noncoding , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , rac GTP-Binding Proteins/genetics
3.
Elife ; 72018 09 20.
Article in English | MEDLINE | ID: mdl-30234487

ABSTRACT

The cereblon modulating agents (CMs) including lenalidomide, pomalidomide and CC-220 repurpose the Cul4-RBX1-DDB1-CRBN (CRL4CRBN) E3 ubiquitin ligase complex to induce the degradation of specific neomorphic substrates via polyubiquitination in conjunction with E2 ubiquitin-conjugating enzymes, which have until now remained elusive. Here we show that the ubiquitin-conjugating enzymes UBE2G1 and UBE2D3 cooperatively promote the K48-linked polyubiquitination of CRL4CRBN neomorphic substrates via a sequential ubiquitination mechanism. Blockade of UBE2G1 diminishes the ubiquitination and degradation of neomorphic substrates, and consequent antitumor activities elicited by all tested CMs. For example, UBE2G1 inactivation significantly attenuated the degradation of myeloma survival factors IKZF1 and IKZF3 induced by lenalidomide and pomalidomide, hence conferring drug resistance. UBE2G1-deficient myeloma cells, however, remained sensitive to a more potent IKZF1/3 degrader CC-220. Collectively, it will be of fundamental interest to explore if loss of UBE2G1 activity is linked to clinical resistance to drugs that hijack the CRL4CRBN to eliminate disease-driving proteins.


Subject(s)
Peptide Hydrolases/metabolism , Proteolysis , Ubiquitin-Conjugating Enzymes/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Cell Line, Tumor , HEK293 Cells , Humans , Ikaros Transcription Factor/metabolism , Substrate Specificity/drug effects , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
4.
J Mich Dent Assoc ; 99(3): 40-2, 68-9, 2017 03.
Article in English | MEDLINE | ID: mdl-30398796

ABSTRACT

Vascular calcification occurs with aging, and several risk factors including diabetes, hyperlipidemia, and disorders of calcium metabolism have been identified. M6nckeberg medial sclerosis (MMS) is the most common variant of medial calcification. M6nckeberg sclerosis can lead to significant adverse cardiovascular outcomes such as arterial stiffness, increased pulse and left ventricular hypertrophy. Here we report two cases of MMS involving facial vasculature, diagnosed incidentally on radiographs during their routine dental evaluation. They appear as convoluted "railroad tracks" patterns of the facial artery calcification. We believe that a better understanding and identification of these calcifications can lead to appropriate patient follow-up with medical providers and interventions to reduce morbidity and mortality by potentially predicting possible cardiovascular events.


Subject(s)
Monckeberg Medial Calcific Sclerosis/diagnostic imaging , Mouth Diseases/diagnostic imaging , Radiography, Panoramic , Diagnosis, Differential , Female , Humans , Male , Middle Aged
5.
Oncotarget ; 7(44): 72356-72372, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27655702

ABSTRACT

Cytogenetic aberrations, such as chromosomal translocations, aneuploidy, and amplifications, are frequently detected in hematological malignancies. For many of the common autosomal aberrations, the mechanisms underlying their roles in cancer development have been well-characterized. On the contrary, although loss of a sex chromosome is observed in a broad range of hematological malignancies, how it cooperates in disease development is less understood. Nevertheless, it has been postulated that tumor suppressor genes reside on the sex chromosomes. Although the X and Y sex chromosomes are highly divergent, the pseudoautosomal regions are homologous between both chromosomes. Here, we review what is currently known about the pseudoautosomal region genes in the hematological system. Additionally, we discuss implications for haploinsufficiency of critical pseudoautosomal region sex chromosome genes, driven by sex chromosome loss, in promoting hematological malignancies. Because mechanistic studies on disease development rely heavily on murine models, we also discuss the challenges and caveats of existing models, and propose alternatives for examining the involvement of pseudoautosomal region genes and loss of a sex chromosome in vivo. With the widespread detection of loss of a sex chromosome in different hematological malignances, the elucidation of the role of pseudoautosomal region genes in the development and progression of these diseases would be invaluable to the field.


Subject(s)
Chromosome Deletion , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , Hematologic Neoplasms/genetics , Pseudoautosomal Regions/genetics , Sex Chromosome Aberrations , Genes, Tumor Suppressor , Humans , Proto-Oncogenes/genetics , RNA, Untranslated/genetics
6.
Mol Cell Biol ; 34(20): 3843-54, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25113560

ABSTRACT

Epidermal growth factor (EGF) receptor (EGFR) has been implicated in tumor development and invasion. Dimerization and autophosphorylation of EGFR are the critical events for EGFR activation. However, the regulation of EGF-dependent and EGF-independent dimerization and phosphorylation of EGFR has not been fully understood. Here, we report that cytoplasmic protein plakophilin-2 (PKP2) is a novel positive regulator of EGFR signaling. PKP2 specifically interacts with EGFR via its N-terminal head domain. Increased PKP2 expression enhances EGF-dependent and EGF-independent EGFR dimerization and phosphorylation. Moreover, PKP2 knockdown reduces EGFR phosphorylation and attenuates EGFR-mediated signal activation, resulting in a significant decrease in proliferation and migration of cancer cells and tumor development. Our results indicate that PKP2 is a novel activator of the EGFR signaling pathway and a potential new drug target for inhibiting tumor growth.


Subject(s)
Carcinogenesis/metabolism , ErbB Receptors/metabolism , Mammary Neoplasms, Experimental/metabolism , Plakophilins/physiology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epidermal Growth Factor/physiology , Female , HEK293 Cells , Humans , Ligands , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Protein Multimerization , Receptor, ErbB-2/metabolism , Signal Transduction , Tumor Burden
7.
Blood ; 124(14): 2203-12, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25150295

ABSTRACT

RUNX1 is a master transcription factor in hematopoiesis and mediates the specification and homeostasis of hematopoietic stem and progenitor cells (HSPCs). Disruptions in RUNX1 are well known to lead to hematologic disease. In this study, we sought to identify and characterize RUNX1 target genes in HSPCs by performing RUNX1 chromatin immunoprecipitation with high-throughput sequencing (ChIP-seq) using a murine HSPC line and complementing this data with our previously described gene expression profiling of primary wild-type and RUNX1-deficient HSPCs (Lineage(-)/cKit(+)/Sca1(+)). From this analysis, we identified and confirmed that Hmga2, a known oncogene, as a direct target of RUNX1. Hmga2 was strongly upregulated in RUNX1-deficient HSPCs, and the promoter of Hmga2 was responsive in a cell-type dependent manner upon coexpression of RUNX1. Conditional Runx1 knockout mice exhibit expansion of their HSPCs and myeloid progenitors as hallmark phenotypes. To further validate and establish that Hmga2 plays a role in inducing HSPC expansion, we generated mouse models of HMGA2 and RUNX1 deficiency. Although mice lacking both factors continued to display higher frequencies of HSPCs, the expansion of myeloid progenitors was effectively rescued. The data presented here establish Hmga2 as a transcriptional target of RUNX1 and a critical regulator of myeloid progenitor expansion.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation , HMGA2 Protein/metabolism , Myeloid Progenitor Cells/cytology , Animals , Binding Sites , Cell Line , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Humans , Jurkat Cells , K562 Cells , Mice , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , Phenotype , Transcription Factors/metabolism , Up-Regulation
8.
Leuk Lymphoma ; 55(4): 884-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23772668

ABSTRACT

The 8;21 translocation is the most common chromosomal aberration occurring in acute myeloid leukemia (AML). This translocation causes expression of the RUNX1-ETO (AML1-ETO) fusion protein, which cooperates with additional mutations in leukemia development. We report here that interferons (IFNs) and IFN-stimulated genes are a group of genes consistently up-regulated by RUNX1-ETO in both human and murine models. RUNX1-ETO-induced up-regulation of IFN-stimulated genes occurs primarily via type I IFN signaling with a requirement for the IFNAR complex. Addition of exogenous IFN in vitro significantly reduces the increase in self-renewal potential induced by both RUNX1-ETO and its leukemogenic splicing isoform RUNX1-ETO9a. Finally, loss of type I IFN signaling via knockout of Ifnar1 significantly accelerates leukemogenesis in a t(8;21) murine model. This demonstrates the role of increased IFN signaling as an important factor inhibiting t(8;21) fusion protein function and leukemia development and supports the use of type I IFNs in the treatment of AML.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic/drug effects , Interferon Type I/pharmacology , Leukemia/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Translocation, Genetic , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Disease Models, Animal , Humans , Leukemia/metabolism , Mice , Mice, Knockout , Oncogene Proteins, Fusion/metabolism , RUNX1 Translocation Partner 1 Protein , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , U937 Cells
9.
Receptors Clin Investig ; 2(1): e485, 2014.
Article in English | MEDLINE | ID: mdl-25995992

ABSTRACT

The oncogenic role of EGFR in many tumors has attracted a great deal of attention in the recent years and initiated the development of several potent EGFR inhibitors, which are used clinically for cancer treatment. However, the current therapeutic inhibition of EGFR signaling is limited to monoclonal antibodies that bind to the EGFR extracellular domain or tyrosine kinase inhibitors that block EGFR kinase activation directly. Despite the great promise of these inhibitors, a certain percentage of patients develop resistance to these therapies, highlighting the necessity for alternative therapeutic strategies based on our most current knowledge of the mechanisms of EGFR signaling. We recently reported that Plakofilin-2 (PKP2) is a novel ligand-independent cytoplasmic activator of EGFR signaling. Here we focus on recent studies demonstrating important roles of intracellular EGFR activators, and propose targeted disruption of these activators as a novel avenue of therapeutic intervention to inhibit EGFR-mediated cancer development.

10.
Cell Cycle ; 11(20): 3792-800, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23070520

ABSTRACT

The DNA damage checkpoint controls cell cycle arrest in response to DNA damage, and activation of this checkpoint is in turn cell cycle-regulated. Rad9, the ortholog of mammalian 53BP1, is essential for this checkpoint response and is phosphorylated by the cyclin-dependent kinase (CDK) in the yeast Saccharomyces cerevisiae. Previous studies suggested that the CDK consensus sites of Rad9 are important for its checkpoint activity. However, the precise CDK sites of Rad9 involved have not been determined. Here we show that CDK consensus sites of Rad9 function in parallel to its BRCT domain toward checkpoint activation, analogous to its fission yeast ortholog Crb2. Unlike Crb2, however, mutation of multiple rather than any individual CDK site of Rad9 is required to completely eliminate its checkpoint activity in vivo. Although Dpb11 interacts with CDK-phosphorylated Rad9, we provide evidence showing that elimination of this interaction does not affect DNA damage checkpoint activation in vivo, suggesting that additional pathway(s) exist. Taken together, these findings suggest that the regulation of Rad9 by CDK and the role of Dpb11 in DNA damage checkpoint activation are more complex than previously suggested. We propose that multiple phosphorylation of Rad9 by CDK may provide a more robust system to allow Rad9 to control cell cycle-dependent DNA damage checkpoint activation.


Subject(s)
Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Cyclin-Dependent Kinases/genetics , DNA Damage , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Sequence Homology, Amino Acid , Signal Transduction
11.
Blood ; 119(13): 3155-63, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22223820

ABSTRACT

The t(8;21)(q22;q22) is common in adult acute myeloid leukemia (AML). The RUNX1-ETO fusion protein that is expressed by this translocation is poorly leukemogenic and requires additional mutations for transformation. Loss of sex chromosome (LOS) is frequently observed in t(8;21) AML. In the present study, to evaluate whether LOS cooperates with t(8;21) in leukemogenesis, we first used a retroviral transduction/transplantation model to express RUNX1-ETO in hematopoietic cells from XO mice. The low frequency of leukemia in these mice suggests that the potentially critical gene for suppression of t(8;21) leukemia in humans is not conserved on mouse sex chromosomes. The gene encoding the GM-CSF receptor α subunit (CSF2RA) is located on X and Y chromosomes in humans but on chromosome 19 in mice. GM-CSF promotes myeloid cell survival, proliferation, and differentiation. To determine whether GM-CSF signaling affects RUNX1-ETO leukemogenesis, hematopoietic stem/progenitor cells that lack GM-CSF signaling were used to express RUNX1-ETO and transplanted into lethally irradiated mice, and a high penetrance of AML was observed in recipients. Furthermore, GM-CSF reduced the replating ability of RUNX1-ETO-expressing cells. These results suggest a possible tumor-suppressor role of GM-CSF in RUNX1-ETO leukemia. Loss of the CSF2RA gene may be a critical mutation explaining the high incidence of LOS associated with the t(8;21)(q22;q22) translocation.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Leukemia, Myeloid, Acute/genetics , Signal Transduction/physiology , Translocation, Genetic , Adult , Animals , Cells, Cultured , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Chromosomes, Mammalian/genetics , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Sex Chromosomes/genetics , Sex Chromosomes/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics
12.
Cell Cycle ; 9(17): 3479-84, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20855976

ABSTRACT

Cells are highly responsive to their environment. One of the main strategies used by cells in signal transduction is protein phosphorylation, a reversible modification that regulates numerous biological processes. Misregulation of phosphorylation-mediated processes is often implicated in many human diseases and cancers. A global and quantitative analysis of protein phosphorylation provides a powerful new approach and has the potential to reveal new insights in signaling pathways. Recent technological advances in high resolution mass spectrometers and multidimensional liquid chromatography, combined with the use of stable isotope labeling of proteins, have led to the application of quantitative phosphoproteomics to study in vivo signal transduction events on a proteome-wide scale. Here we review recent advancements in quantitative phosphoproteomic technologies, discuss their potentials and identify areas for future development. A key objective of proteomic technology is its application to addressing biological questions. We will therefore describe how current quantitative phosphoproteomic technology can be used to study the molecular basis of phosphorylation events in the DNA damage response.


Subject(s)
DNA Repair , Proteomics , DNA Damage , Humans , Mass Spectrometry , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphorylation , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...