Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Natl Cancer Inst ; 115(7): 838-852, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37040084

ABSTRACT

BACKGROUND: Adenoid cystic carcinoma (ACC) is a lethal malignancy of exocrine glands, characterized by the coexistence within tumor tissues of 2 distinct populations of cancer cells, phenotypically similar to the myoepithelial and ductal lineages of normal salivary epithelia. The developmental relationship linking these 2 cell types, and their differential vulnerability to antitumor treatments, remains unknown. METHODS: Using single-cell RNA sequencing, we identified cell-surface markers (CD49f, KIT) that enabled the differential purification of myoepithelial-like (CD49fhigh/KITneg) and ductal-like (CD49flow/KIT+) cells from patient-derived xenografts (PDXs) of human ACCs. Using prospective xenotransplantation experiments, we compared the tumor-initiating capacity of the 2 cell types and tested whether one could differentiate into the other. Finally, we searched for signaling pathways with differential activation between the 2 cell types and tested their role as lineage-specific therapeutic targets. RESULTS: Myoepithelial-like cells displayed higher tumorigenicity than ductal-like cells and acted as their progenitors. Myoepithelial-like and ductal-like cells displayed differential expression of genes encoding for suppressors and activators of retinoic acid signaling, respectively. Agonists of retinoic acid receptor (RAR) or retinoid X receptor (RXR) signaling (all-trans retinoic acid, bexarotene) promoted myoepithelial-to-ductal differentiation, whereas suppression of RAR/RXR signaling with a dominant-negative RAR construct abrogated it. Inverse agonists of RAR/RXR signaling (BMS493, AGN193109) displayed selective toxicity against ductal-like cells and in vivo antitumor activity against PDX models of human ACC. CONCLUSIONS: In human ACCs, myoepithelial-like cells act as progenitors of ductal-like cells, and myoepithelial-to-ductal differentiation is promoted by RAR/RXR signaling. Suppression of RAR/RXR signaling is lethal to ductal-like cells and represents a new therapeutic approach against human ACCs.


Subject(s)
Antineoplastic Agents , Carcinoma, Adenoid Cystic , Receptors, Retinoic Acid , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Adenoid Cystic/drug therapy , Drug Inverse Agonism , Prospective Studies , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Tretinoin
2.
Nat Commun ; 14(1): 2290, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37085479

ABSTRACT

Tissue homeostasis is maintained after stress by engaging and activating the hematopoietic stem and progenitor compartments in the blood. Hematopoietic stem cells (HSCs) are essential for long-term repopulation after secondary transplantation. Here, using a conditional knockout mouse model, we revealed that the RNA-binding protein SYNCRIP is required for maintenance of blood homeostasis especially after regenerative stress due to defects in HSCs and progenitors. Mechanistically, we find that SYNCRIP loss results in a failure to maintain proteome homeostasis that is essential for HSC maintenance. SYNCRIP depletion results in increased protein synthesis, a dysregulated epichaperome, an accumulation of misfolded proteins and induces endoplasmic reticulum stress. Additionally, we find that SYNCRIP is required for translation of CDC42 RHO-GTPase, and loss of SYNCRIP results in defects in polarity, asymmetric segregation, and dilution of unfolded proteins. Forced expression of CDC42 recovers polarity and in vitro replating activities of HSCs. Taken together, we uncovered a post-transcriptional regulatory program that safeguards HSC self-renewal capacity and blood homeostasis.


Subject(s)
Hematopoietic Stem Cells , Heterogeneous-Nuclear Ribonucleoproteins , Proteostasis , Animals , Mice , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Mice, Knockout , Proteostasis/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Cancer Cell ; 41(4): 726-739.e11, 2023 04 10.
Article in English | MEDLINE | ID: mdl-36898380

ABSTRACT

Acute myeloid leukemia (AML) is a hematologic malignancy for which several epigenetic regulators have been identified as therapeutic targets. Here we report the development of cereblon-dependent degraders of IKZF2 and casein kinase 1α (CK1α), termed DEG-35 and DEG-77. We utilized a structure-guided approach to develop DEG-35 as a nanomolar degrader of IKZF2, a hematopoietic-specific transcription factor that contributes to myeloid leukemogenesis. DEG-35 possesses additional substrate specificity for the therapeutically relevant target CK1α, which was identified through unbiased proteomics and a PRISM screen assay. Degradation of IKZF2 and CK1α blocks cell growth and induces myeloid differentiation in AML cells through CK1α-p53- and IKZF2-dependent pathways. Target degradation by DEG-35 or a more soluble analog, DEG-77, delays leukemia progression in murine and human AML mouse models. Overall, we provide a strategy for multitargeted degradation of IKZF2 and CK1α to enhance efficacy against AML that may be expanded to additional targets and indications.


Subject(s)
Casein Kinase Ialpha , Leukemia, Myeloid, Acute , Animals , Humans , Mice , Casein Kinase Ialpha/genetics , Casein Kinase Ialpha/metabolism , Hematopoiesis , Ikaros Transcription Factor/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Transcription Factors
4.
Clin Cancer Res ; 29(5): 943-956, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36537918

ABSTRACT

PURPOSE: The Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) polycythemia vera, essential thrombocythemia, and primary myelofibrosis are characterized by JAK/STAT pathway activation. JAK inhibitors are approved for MPN treatment, but persistence has been observed, due to JAK/STAT reactivation. EXPERIMENTAL DESIGN: Using MPN patient samples, JAK2-mutated cell lines, and MPN mouse models, we examined both the efficacy and mechanism by which crizotinib, the ALK/MET/RON/ROS1 inhibitor approved for the treatment of non-small cell lung cancer, alters MPN cell proliferation and JAK/STAT activation. RESULTS: We found that crizotinib suppresses proliferation and activation of JAK/STAT signaling, and decreases the disease burden in the JAK2V617F mouse model of MPN. Furthermore, we found that crizotinib could overcome JAK inhibitor persistence to ruxolitinib. Interestingly, phosphorylation of the crizotinib target RON kinase was enhanced in ruxolitinib-persistent cells. We show that phospho-JAK2 and phospho-RON can physically interact to sustain JAK/STAT signaling, and that the combination of crizotinib and ruxolitinib disrupts this interaction. Furthermore, RON knockdown suppresses proliferation and activation of JAK/STAT signaling in JAK2-mutated cells, and RON deletion in a JAK2V617F mouse MPN model decreases the disease burden. We also observed RON hyperactivation in MPN patient cells, suggesting that RON may be an important target of crizotinib in MPN. CONCLUSIONS: In summary, we demonstrate that crizotinib has preclinical efficacy in MPN patient cells, JAK2-mutated cell lines, and a JAK2-mutated mouse model, and that the combination of crizotinib with JAK inhibitors suppresses JAK inhibitor persistence. Our work suggests that crizotinib should be investigated for the treatment of patients with MPN.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Janus Kinase Inhibitors , Lung Neoplasms , Myeloproliferative Disorders , Animals , Mice , Janus Kinase Inhibitors/therapeutic use , Crizotinib/pharmacology , Crizotinib/therapeutic use , Janus Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Signal Transduction , STAT Transcription Factors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Janus Kinase 2/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mutation
5.
Nat Cancer ; 2: 741-757, 2021 07.
Article in English | MEDLINE | ID: mdl-34458856

ABSTRACT

RNA binding proteins (RBPs) are key arbiters of post-transcriptional regulation and are found to be found dysregulated in hematological malignancies. Here, we identify the RBP RBMX and its retrogene RBMXL1 to be required for murine and human myeloid leukemogenesis. RBMX/L1 are overexpressed in acute myeloid leukemia (AML) primary patients compared to healthy individuals, and RBMX/L1 loss delayed leukemia development. RBMX/L1 loss lead to significant changes in chromatin accessibility, as well as chromosomal breaks and gaps. We found that RBMX/L1 directly bind to mRNAs, affect transcription of multiple loci, including CBX5 (HP1α), and control the nascent transcription of the CBX5 locus. Forced CBX5 expression rescued the RBMX/L1 depletion effects on cell growth and apoptosis. Overall, we determine that RBMX/L1 control leukemia cell survival by regulating chromatin state through their downstream target CBX5. These findings identify a mechanism for RBPs directly promoting transcription and suggest RBMX/L1, as well as CBX5, as potential therapeutic targets in myeloid malignancies.


Subject(s)
Chromatin , Leukemia, Myeloid, Acute , Animals , Chromatin/genetics , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , RNA-Binding Proteins/genetics , Transcription Factors/genetics
6.
Cell Metab ; 33(1): 145-159.e6, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33357456

ABSTRACT

A significant increase in dietary fructose consumption has been implicated as a potential driver of cancer. Metabolic adaptation of cancer cells to utilize fructose confers advantages for their malignant growth, but compelling therapeutic targets have not been identified. Here, we show that fructose metabolism of leukemic cells can be inhibited by targeting the de novo serine synthesis pathway (SSP). Leukemic cells, unlike their normal counterparts, become significantly dependent on the SSP in fructose-rich conditions as compared to glucose-rich conditions. This metabolic program is mediated by the ratio of redox cofactors, NAD+/NADH, and the increased SSP flux is beneficial for generating alpha-ketoglutarate from glutamine, which allows leukemic cells to proliferate even in the absence of glucose. Inhibition of PHGDH, a rate-limiting enzyme in the SSP, dramatically reduces leukemia engraftment in mice in the presence of high fructose, confirming the essential role of the SSP in the metabolic plasticity of leukemic cells.


Subject(s)
Fructose/metabolism , Leukemia, Myeloid, Acute/metabolism , Serine/biosynthesis , Animals , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Congenic , Mice, Inbred NOD , Tumor Cells, Cultured
7.
Nat Commun ; 11(1): 2026, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332729

ABSTRACT

The cell-context dependency for RNA binding proteins (RBPs) mediated control of stem cell fate remains to be defined. Here we adapt the HyperTRIBE method using an RBP fused to a Drosophila RNA editing enzyme (ADAR) to globally map the mRNA targets of the RBP MSI2 in mammalian adult normal and malignant stem cells. We reveal a unique MUSASHI-2 (MSI2) mRNA binding network in hematopoietic stem cells that changes during transition to multipotent progenitors. Additionally, we discover a significant increase in RNA binding activity of MSI2 in leukemic stem cells compared with normal hematopoietic stem and progenitor cells, resulting in selective regulation of MSI2's oncogenic targets. This provides a basis for MSI2 increased dependency in leukemia cells compared to normal cells. Moreover, our study provides a way to measure RBP function in rare cells and suggests that RBPs can achieve differential binding activity during cell state transition independent of gene expression.


Subject(s)
Cell Differentiation/genetics , Hematopoietic Stem Cells/pathology , Leukemia/genetics , Neoplastic Stem Cells/pathology , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Animals , Binding Sites/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Gene Expression Regulation, Leukemic , Gene Regulatory Networks , HEK293 Cells , Humans , Leukemia/blood , Leukemia/pathology , Mice , Mice, Knockout , Primary Cell Culture , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Seq , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
8.
Cell Rep ; 28(7): 1703-1716.e6, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412241

ABSTRACT

Stem cells balance cellular fates through asymmetric and symmetric divisions in order to self-renew or to generate downstream progenitors. Symmetric commitment divisions in stem cells are required for rapid regeneration during tissue damage and stress. The control of symmetric commitment remains poorly defined. Using single-cell RNA sequencing (scRNA-seq) in combination with transcriptomic profiling of HSPCs (hematopoietic stem and progenitor cells) from control and m6A methyltransferase Mettl3 conditional knockout mice, we found that m6A-deficient hematopoietic stem cells (HSCs) fail to symmetrically differentiate. Dividing HSCs are expanded and are blocked in an intermediate state that molecularly and functionally resembles multipotent progenitors. Mechanistically, RNA methylation controls Myc mRNA abundance in differentiating HSCs. We identified MYC as a marker for HSC asymmetric and symmetric commitment. Overall, our results indicate that RNA methylation controls symmetric commitment and cell identity of HSCs and may provide a general mechanism for how stem cells regulate differentiation fate choice.


Subject(s)
Cell Differentiation , Cell Lineage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Methyltransferases/physiology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Female , Hematopoietic Stem Cells/metabolism , High-Throughput Nucleotide Sequencing , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-myc/genetics , RNA Stability , Single-Cell Analysis
9.
Nat Commun ; 10(1): 2691, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217428

ABSTRACT

The MUSASHI (MSI) family of RNA binding proteins (MSI1 and MSI2) contribute to a wide spectrum of cancers including acute myeloid leukemia. We find that the small molecule Ro 08-2750 (Ro) binds directly and selectively to MSI2 and competes for its RNA binding in biochemical assays. Ro treatment in mouse and human myeloid leukemia cells results in an increase in differentiation and apoptosis, inhibition of known MSI-targets, and a shared global gene expression signature similar to shRNA depletion of MSI2. Ro demonstrates in vivo inhibition of c-MYC and reduces disease burden in a murine AML leukemia model. Thus, we identify a small molecule that targets MSI's oncogenic activity. Our study provides a framework for targeting RNA binding proteins in cancer.


Subject(s)
Gene Expression Regulation, Leukemic/drug effects , Leukemia, Experimental/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Pteridines/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Flavins , Gene Expression Profiling , Humans , Leukemia, Experimental/blood , Leukemia, Myeloid, Acute/blood , Male , Mice , Primary Cell Culture , Proto-Oncogene Proteins c-myc/metabolism , Pteridines/therapeutic use , RNA/metabolism , RNA Recognition Motif/drug effects , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcriptome/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...