Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
Cell ; 140(5): 652-65, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20211135

ABSTRACT

MicroRNAs and heterogeneous ribonucleoproteins (hnRNPs) are posttranscriptional gene regulators that bind mRNA in a sequence-specific manner. Here, we report that loss of miR-328 occurs in blast crisis chronic myelogenous leukemia (CML-BC) in a BCR/ABL dose- and kinase-dependent manner through the MAPK-hnRNP E2 pathway. Restoration of miR-328 expression rescues differentiation and impairs survival of leukemic blasts by simultaneously interacting with the translational regulator poly(rC)-binding protein hnRNP E2 and with the mRNA encoding the survival factor PIM1, respectively. The interaction with hnRNP E2 is independent of the microRNA's seed sequence and it leads to release of CEBPA mRNA from hnRNP E2-mediated translational inhibition. Altogether, these data reveal the dual ability of a microRNA to control cell fate both through base pairing with mRNA targets and through a decoy activity that interferes with the function of regulatory proteins.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MicroRNAs/metabolism , Animals , Blast Crisis , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Proto-Oncogene Proteins c-pim-1/metabolism , RNA-Induced Silencing Complex/metabolism
2.
Haematologica ; 104(12): 2400-2409, 2019 12.
Article in English | MEDLINE | ID: mdl-31073075

ABSTRACT

There are no validated molecular biomarkers to identify newly-diagnosed individuals with chronic-phase chronic myeloid leukemia likely to respond poorly to imatinib and who might benefit from first-line treatment with a more potent second-generation tyrosine kinase inhibitor. Our inability to predict these 'high-risk' individuals reflects the poorly understood heterogeneity of the disease. To investigate the potential of genetic variants in epigenetic modifiers as biomarkers at diagnosis, we used Ion Torrent next-generation sequencing of 71 candidate genes for predicting response to tyrosine kinase inhibitors and probability of disease progression. A total of 124 subjects with newly-diagnosed chronic-phase chronic myeloid leukemia began with imatinib (n=62) or second-generation tyrosine kinase inhibitors (n=62) and were classified as responders or non-responders based on the BCRABL1 transcript levels within the first year and the European LeukemiaNet criteria for failure. Somatic variants affecting 21 genes (e.g. ASXL1, IKZF1, DNMT3A, CREBBP) were detected in 30% of subjects, most of whom were non-responders (41% non-responders, 18% responders to imatinib, 38% non-responders, 25% responders to second-generation tyrosine kinase inhibitors). The presence of variants predicted the rate of achieving a major molecular response, event-free survival, progression-free survival and chronic myeloid leukemia-related survival in the imatinib but not the second-generation tyrosine kinase inhibitors cohort. Rare germline variants had no prognostic significance irrespective of treatment while some pre-leukemia variants suggest a multi-step development of chronic myeloid leukemia. Our data suggest that identification of somatic variants at diagnosis facilitates stratification into imatinib responders/non-responders, thereby allowing earlier use of second-generation tyrosine kinase inhibitors, which, in turn, may overcome the negative impact of such variants on disease progression.


Subject(s)
Biomarkers, Tumor/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutation , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Male , Middle Aged , Retrospective Studies , Survival Rate , Treatment Failure , Young Adult
3.
Haematologica ; 103(12): 2016-2025, 2018 12.
Article in English | MEDLINE | ID: mdl-30049824

ABSTRACT

The fusion oncoprotein BCR-ABL1 exhibits aberrant tyrosine kinase activity and it has been proposed that it deregulates signaling networks involving both transcription factors and non-coding microRNAs that result in chronic myeloid leukemia (CML). Previously, microRNA expression profiling showed deregulated expression of miR-150 and miR-155 in CML. In this study, we placed these findings into the broader context of the MYC/miR-150/MYB/miR-155/PU.1 oncogenic network. We propose that up-regulated MYC and miR-155 in CD34+ leukemic stem and progenitor cells, in concert with BCR-ABL1, impair the molecular mechanisms of myeloid differentiation associated with low miR-150 and PU.1 levels. We revealed that MYC directly occupied the -11.7 kb and -0.35 kb regulatory regions in the MIR150 gene. MYC occupancy was markedly increased through BCR-ABL1 activity, causing inhibition of MIR150 gene expression in CML CD34+ and CD34- cells. Furthermore, we found an association between reduced miR-150 levels in CML blast cells and their resistance to tyrosine kinase inhibitors (TKIs). Although TKIs successfully disrupted BCR-ABL1 kinase activity in proliferating CML cells, this treatment did not efficiently target quiescent leukemic stem cells. The study presents new evidence regarding the MYC/miR-150/MYB/miR-155/PU.1 leukemic network established by aberrant BCR-ABL1 activity. The key connecting nodes of this network may serve as potential druggable targets to overcome resistance of CML stem and progenitor cells.


Subject(s)
Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Genes, myc/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MicroRNAs/genetics , Adult , Aged , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology
4.
Hematol Oncol ; 36(5): 740-748, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30074634

ABSTRACT

Following the 47th American Society of Hematology Meeting in 2005, the late John Goldman and Tariq Mughal commenced a conference, the 1st Post-ASH Workshop, which brought together clinicians and scientists, to accelerate the adoption of new therapies for patients with myeloproliferative neoplasms (MPNs). The concept began with recognition of the CML success story following imatinib therapy, the discovery of JAK2V617F , and the demonstration that BCR-ABL1-negative MPNs are driven by abnormal JAK2 activation. This review is based on the presentations and deliberations at the XIIth Post-ASH Workshop on BCR-ABL1 positive and negative MPNs that took place on December 12 to 13, 2017, in Atlanta, Georgia, immediately following the 59th American Society of Hematology Meeting. We have selected some of the translational research and clinical topics, rather than an account of the proceedings. We discuss the role of immunotherapy in MPNs and the impact of the mutational landscape on TKI treatment in CML. We also consider how we might reduce TKI cardiovascular side effects, the potential role of nutrition as adjunctive nonpharmacologic intervention to reduce chronic inflammation in MPNs, and novel investigational therapies for MPNs.


Subject(s)
Hematologic Neoplasms , Immunotherapy/methods , Myeloproliferative Disorders , Precision Medicine/methods , Amino Acid Substitution , Fusion Proteins, bcr-abl , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Humans , Janus Kinase 2 , Mutation, Missense , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/therapy , Translational Research, Biomedical
5.
Blood ; 123(18): 2826-37, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24668492

ABSTRACT

Acute myeloid leukemia (AML) is sustained by small populations of leukemia stem cells (LSCs) that can resist available treatments and represent important barriers to cure. Although previous studies have shown increased signal transducer and activator of transcription (STAT)3 and STAT5 phosphorylation in AML leukemic blasts, the role of Janus kinase (JAK) signaling in primary AML compared with normal stem cells has not been directly evaluated. We show here that JAK/STAT signaling is increased in LSCs, particularly from high-risk AML. JAK2 inhibition using small molecule inhibitors or interference RNA reduced growth of AML LSCs while sparing normal stem cells both in vitro and in vivo. Increased JAK/STAT activity was associated with increased expression and altered signaling through growth factor receptors in AML LSCs, including receptor tyrosine kinase c-KIT and FMS-related tyrosine kinase 3 (FLT3). Inhibition of c-KIT and FLT3 expression significantly inhibited JAK/STAT signaling in AML LSCs, and JAK inhibitors effectively inhibited FLT3-mutated AML LSCs. Our results indicate that JAK/STAT signaling represents an important signaling mechanism supporting AML LSC growth and survival. These studies support continued evaluation of strategies for JAK/STAT inhibition for therapeutic targeting of AML LSCs.


Subject(s)
Janus Kinase 2/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction , Animals , Antigens, CD34/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Leukemic , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinases/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Neoplastic Stem Cells/drug effects , Phenotype , Phosphorylation , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA Interference , Receptors, Growth Factor/genetics , STAT Transcription Factors/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
PLoS Genet ; 9(3): e1003311, 2013.
Article in English | MEDLINE | ID: mdl-23505378

ABSTRACT

MicroRNAs (miRNAs), single-stranded non-coding RNAs, influence myriad biological processes that can contribute to cancer. Although tumor-suppressive and oncogenic functions have been characterized for some miRNAs, the majority of microRNAs have not been investigated for their ability to promote and modulate tumorigenesis. Here, we established that the miR-191/425 cluster is transcriptionally dependent on the host gene, DALRD3, and that the hormone 17ß-estradiol (estrogen or E2) controls expression of both miR-191/425 and DALRD3. MiR-191/425 locus characterization revealed that the recruitment of estrogen receptor α (ERα) to the regulatory region of the miR-191/425-DALRD3 unit resulted in the accumulation of miR-191 and miR-425 and subsequent decrease in DALRD3 expression levels. We demonstrated that miR-191 protects ERα positive breast cancer cells from hormone starvation-induced apoptosis through the suppression of tumor-suppressor EGR1. Furthermore, enforced expression of the miR-191/425 cluster in aggressive breast cancer cells altered global gene expression profiles and enabled us to identify important tumor promoting genes, including SATB1, CCND2, and FSCN1, as targets of miR-191 and miR-425. Finally, in vitro and in vivo experiments demonstrated that miR-191 and miR-425 reduced proliferation, impaired tumorigenesis and metastasis, and increased expression of epithelial markers in aggressive breast cancer cells. Our data provide compelling evidence for the transcriptional regulation of the miR-191/425 cluster and for its context-specific biological determinants in breast cancers. Importantly, we demonstrated that the miR-191/425 cluster, by reducing the expression of an extensive network of genes, has a fundamental impact on cancer initiation and progression of breast cancer cells.


Subject(s)
Breast Neoplasms , Early Growth Response Protein 1 , Estrogen Receptor alpha , MicroRNAs , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism
7.
Blood ; 122(11): 1923-34, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23926298

ABSTRACT

FTY720 (Fingolimod, Gilenya) is a sphingosine analog used as an immunosuppressant in multiple sclerosis patients. FTY720 is also a potent protein phosphatase 2A (PP2A)-activating drug (PAD). PP2A is a tumor suppressor found inactivated in different types of cancer. We show here that PP2A is inactive in polycythemia vera (PV) and other myeloproliferative neoplasms characterized by the expression of the transforming Jak2(V617F) oncogene. PP2A inactivation occurs in a Jak2(V617F) dose/kinase-dependent manner through the PI-3Kγ-PKC-induced phosphorylation of the PP2A inhibitor SET. Genetic or PAD-mediated PP2A reactivation induces Jak2(V617F) inactivation/downregulation and impairs clonogenic potential of Jak2(V617F) cell lines and PV but not normal CD34(+) progenitors. Likewise, FTY720 decreases leukemic allelic burden, reduces splenomegaly, and significantly increases survival of Jak2(V617F) leukemic mice without adverse effects. Mechanistically, we show that in Jak2(V617F) cells, FTY720 antileukemic activity requires neither FTY720 phosphorylation (FTY720-P) nor SET dimerization or ceramide induction but depends on interaction with SET K209. Moreover, we show that Jak2(V617F) also utilizes an alternative sphingosine kinase-1-mediated pathway to inhibit PP2A and that FTY720-P, acting as a sphingosine-1-phosphate-receptor-1 agonist, elicits signals leading to the Jak2-PI-3Kγ-PKC-SET-mediated PP2A inhibition. Thus, PADs (eg, FTY720) represent suitable therapeutic alternatives for Jak2(V617F) MPNs.


Subject(s)
Janus Kinase 2/metabolism , Leukemia/drug therapy , Propylene Glycols/pharmacology , Protein Phosphatase 2/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase , DNA-Binding Proteins , Enzyme Activation/drug effects , Fingolimod Hydrochloride , Histone Chaperones , Humans , Immunoblotting , Immunosuppressive Agents/pharmacology , Janus Kinase 2/genetics , Kaplan-Meier Estimate , Leukemia/genetics , Leukemia/pathology , Mice , Mice, SCID , Mutation , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Phosphatase 2/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sphingosine/pharmacology , Treatment Outcome
8.
Blood ; 122(17): 3034-44, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-23970380

ABSTRACT

As tyrosine kinase inhibitors (TKIs) fail to induce long-term response in blast crisis chronic myelogenous leukemia (CML-BC) and Philadelphia chromosome-positive (Ph(+)) acute lymphoblastic leukemia (ALL), novel therapies targeting leukemia-dysregulated pathways are necessary. Exportin-1 (XPO1), also known as chromosome maintenance protein 1, regulates cell growth and differentiation by controlling the nucleocytoplasmic trafficking of proteins and RNAs, some of which are aberrantly modulated in BCR-ABL1(+) leukemias. Using CD34(+) progenitors from CML, B-ALL, and healthy individuals, we found that XPO1 expression was markedly increased, mostly in a TKI-sensitive manner, in CML-BC and Ph(+) B-ALL. Notably, XPO1 was also elevated in Ph(-) B-ALL. Moreover, the clinically relevant XPO1 inhibitor KPT-330 strongly triggered apoptosis and impaired the clonogenic potential of leukemic, but not normal, CD34(+) progenitors, and increased survival of BCR-ABL1(+) mice, 50% of which remained alive and, mostly, became BCR-ABL1 negative. Moreover, KPT-330 compassionate use in a patient with TKI-resistant CML undergoing disease progression significantly reduced white blood cell count, blast cells, splenomegaly, lactate dehydrogenase levels, and bone pain. Mechanistically, KPT-330 altered the subcellular localization of leukemia-regulated factors including RNA-binding heterogeneous nuclear ribonucleoprotein A1 and the oncogene SET, thereby inducing reactivation of protein phosphatase 2A tumor suppressor and inhibition of BCR-ABL1 in CML-BC cells. Because XPO1 is important for leukemic cell survival, KPT-330 may represent an alternative therapy for TKI-refractory Ph(+) leukemias.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Hydrazines/pharmacology , Karyopherins/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/pharmacology , Adult , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , DNA-Binding Proteins , Drug Evaluation, Preclinical , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/genetics , Histone Chaperones/metabolism , Humans , Karyopherins/genetics , Karyopherins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Transport , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Ribonucleoproteins/antagonists & inhibitors , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Exportin 1 Protein
9.
Blood ; 121(1): 159-69, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-23100311

ABSTRACT

Recently, we showed that increased miR-181a expression was associated with improved outcomes in cytogenetically normal acute myeloid leukemia (CN-AML). Interestingly, miR-181a expression was increased in CN-AML patients harboring CEBPA mutations, which are usually biallelic and associate with better prognosis. CEBPA encodes the C/EBPα transcription factor. We demonstrate here that the presence of N-terminal CEBPA mutations and miR-181a expression are linked. Indeed, the truncated C/EBPα-p30 isoform, which is produced from the N-terminal mutant CEBPA gene or from the differential translation of wild-type CEBPA mRNA and is commonly believed to have no transactivation activity, binds to the miR-181a-1 promoter and up-regulates the microRNA expression. Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPα-p30 isoform, resulting in higher miR-181a levels. In xenograft mouse models, ectopic miR-181a expression inhibits tumor growth. Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. This regulatory pathway may explain an increased sensitivity to apoptosis-inducing chemotherapy in subsets of AML patients. Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPα-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Gene Expression Regulation, Leukemic/drug effects , Immunologic Factors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , MicroRNAs/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/biosynthesis , Thalidomide/analogs & derivatives , Adult , Animals , Antimetabolites, Antineoplastic/pharmacology , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/transplantation , Cytarabine/pharmacology , Frameshift Mutation , Humans , Immunologic Factors/therapeutic use , K562 Cells , Lenalidomide , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Neoplasm Proteins/genetics , Point Mutation , Promoter Regions, Genetic/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Structure, Tertiary , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/physiology , Thalidomide/pharmacology , Thalidomide/therapeutic use , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
10.
Cancer Cell ; 12(5): 467-78, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17996650

ABSTRACT

Chronic myelogenous leukemia (CML) is a clonal myeloproliferative disease (MPD) initiated by expression of the p210-BCR-ABL fusion protein. We demonstrate in a murine model of p210-BCR-ABL-induced MPD that gene targeting of Rac1 and Rac2 significantly delays or abrogates disease development. Attenuation of the disease phenotype is associated with severely diminished p210-BCR-ABL-induced downstream signaling in primary hematopoietic cells. We utilize NSC23766, a small molecule antagonist of Rac activation, to validate biochemically and functionally Rac as a molecular target in both a relevant animal model and in primary human CML cells in vitro and in a xenograft model in vivo, including in Imatinib-resistant p210-BCR-ABL disease. These data demonstrate that Rac is an additional therapeutic target in p210-BCR-ABL-mediated MPD.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , rac GTP-Binding Proteins/physiology , Aminoquinolines/pharmacology , Animals , Antigens, CD34/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Mice , Myeloproliferative Disorders/therapy , Neoplasm Transplantation , Phenotype , Pyrimidines/pharmacology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , RAC2 GTP-Binding Protein
11.
Proc Natl Acad Sci U S A ; 109(49): 20047-52, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23169640

ABSTRACT

Multiple studies have established that microRNAs (miRNAs) are involved in the initiation and progression of cancer. Notably, miR-155 is one of the most overexpressed miRNAs in several solid and hematological malignancies. Ectopic miR-155 expression in mice B cells (Eµ-miR-155 transgenic mice) has been shown to induce pre-B-cell proliferation followed by high-grade lymphoma/leukemia. Loss of miR-155 in mice resulted in impaired immunity due to defective T-cell-mediated immune response. Here we provide a mechanistic insight into miR-155-induced leukemogenesis in the Eµ-miR-155 mouse model through genome-wide transcriptome analysis of naïve B cells and target studies. We found that a key transcriptional repressor and proto-oncogene, Bcl6 is significantly down-regulated in Eµ-miR-155 mice. The reduction of Bcl6 subsequently leads to de-repression of some of the known Bcl6 targets like inhibitor of differentiation (Id2), interleukin-6 (IL6), cMyc, Cyclin D1, and Mip1α/ccl3, all of which promote cell survival and proliferation. We show that Bcl6 is indirectly regulated by miR-155 through Mxd1/Mad1 up-regulation. Interestingly, we found that miR-155 directly targets HDAC4, a corepressor partner of BCL6. Furthermore, ectopic expression of HDAC4 in human-activated B-cell-type diffuse large B-cell lymphoma (DLBCL) cells results in reduced miR-155-induced proliferation, clonogenic potential, and increased apoptosis. Meta-analysis of the diffuse large B-cell lymphoma patient microarray data showed that miR-155 expression is inversely correlated with Bcl6 and Hdac4. Hence this study provides a better understanding of how miR-155 causes disruption of the BCL6 transcriptional machinery that leads to up-regulation of the survival and proliferation genes in miR-155-induced leukemias.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic/immunology , Histone Deacetylases/metabolism , Leukemia, Lymphoid/etiology , MicroRNAs/pharmacology , Proto-Oncogene Proteins c-bcl-6/metabolism , Transcription, Genetic/drug effects , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line , Cyclin D1/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunoblotting , Inhibitor of Differentiation Protein 2/metabolism , Interleukin-6/metabolism , Leukemia, Lymphoid/immunology , Leukemia, Lymphoid/metabolism , Luciferases , Mice , Mice, Transgenic , MicroRNAs/genetics , Microarray Analysis , Proto-Oncogene Mas , Real-Time Polymerase Chain Reaction , Repressor Proteins/metabolism , Signal Transduction/physiology
12.
Proc Natl Acad Sci U S A ; 109(31): E2110-6, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22753494

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs, 19-24 nucleotides in length, that regulate gene expression and are expressed aberrantly in most types of cancer. MiRNAs also have been detected in the blood of cancer patients and can serve as circulating biomarkers. It has been shown that secreted miRNAs within exosomes can be transferred from cell to cell and can regulate gene expression in the receiving cells by canonical binding to their target messenger RNAs. Here we show that tumor-secreted miR-21 and miR-29a also can function by another mechanism, by binding as ligands to receptors of the Toll-like receptor (TLR) family, murine TLR7 and human TLR8, in immune cells, triggering a TLR-mediated prometastatic inflammatory response that ultimately may lead to tumor growth and metastasis. Thus, by acting as paracrine agonists of TLRs, secreted miRNAs are key regulators of the tumor microenvironment. This mechanism of action of miRNAs is implicated in tumor-immune system communication and is important in tumor growth and spread, thus representing a possible target for cancer treatment.


Subject(s)
Membrane Glycoproteins/metabolism , MicroRNAs/blood , Neoplasms/blood , RNA, Neoplasm/blood , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Animals , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Paracrine Communication/genetics , RNA, Neoplasm/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics
13.
Blood ; 119(20): 4786-97, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22408260

ABSTRACT

Acute graft-versus-host disease (aGVHD) remains a major complication of allogeneic hematopoietic stem cell transplant (alloHSCT), underscoring the need to further elucidate its mechanisms and develop novel treatments. Based on recent observations that microRNA-155 (miR-155) is up-regulated during T-cell activation, we hypothesized that miR-155 is involved in the modulation of aGVHD. Here we show that miR-155 expression was up-regulated in T cells from mice developing aGVHD after alloHSCT. Mice receiving miR-155-deficient donor lymphocytes had markedly reduced lethal aGVHD, whereas lethal aGVHD developed rapidly in mice recipients of miR-155 overexpressing T cells. Blocking miR-155 expression using a synthetic anti-miR-155 after alloHSCT decreased aGVHD severity and prolonged survival in mice. Finally, miR-155 up-regulation was shown in specimens from patients with pathologic evidence of intestinal aGVHD. Altogether, our data indicate a role for miR-155 in the regulation of GVHD and point to miR-155 as a novel target for therapeutic intervention in this disease.


Subject(s)
Graft vs Host Disease/genetics , MicroRNAs/physiology , Acute Disease , Animals , Cells, Cultured , Female , Gene Expression Regulation/genetics , Genetic Therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Humans , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Spleen/cytology , Spleen/metabolism , Spleen/transplantation , T-Lymphocytes/metabolism
14.
Blood ; 119(25): 6025-31, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22566605

ABSTRACT

We recently reported promising clinical activity for a 10-day regimen of decitabine in older AML patients; high miR-29b expression associated with clinical response. Subsequent preclinical studies with bortezomib in AML cells have shown drug-induced miR-29b up-regulation, resulting in loss of transcriptional activation for several genes relevant to myeloid leukemogenesis, including DNA methyltransferases and receptor tyrosine kinases. Thus, a phase 1 trial of bortezomib and decitabine was developed. Nineteen poor-risk AML patients (median age 70 years; range, 32-84 years) enrolled. Induction with decitabine (20 mg/m(2) intravenously on days 1-10) plus bortezomib (escalated up to the target 1.3 mg/m(2) on days 5, 8, 12, and 15) was tolerable, but bortezomib-related neuropathy developed after repetitive cycles. Of previously untreated patients (age ≥ 65 years), 5 of 10 had CR (complete remission, n = 4) or incomplete CR (CRi, n = 1); 7 of 19 overall had CR/CRi. Pharmacodynamic analysis showed FLT3 down-regulation on day 26 of cycle 1 (P = .02). Additional mechanistic studies showed that FLT3 down-regulation was due to bortezomib-induced miR-29b up-regulation; this led to SP1 down-regulation and destruction of the SP1/NF-κB complex that transactivated FLT3. This study demonstrates the feasibility and preliminary clinical activity of decitabine plus bortezomib in AML and identifies FLT3 as a novel pharmacodynamic end point for future trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/analogs & derivatives , Boronic Acids/administration & dosage , Boronic Acids/pharmacokinetics , Leukemia, Myeloid, Acute/drug therapy , Pyrazines/administration & dosage , Pyrazines/pharmacokinetics , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azacitidine/administration & dosage , Azacitidine/pharmacokinetics , Azacitidine/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Decitabine , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Leukemic/drug effects , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Pyrazines/pharmacology , Treatment Outcome , Validation Studies as Topic
15.
Proc Natl Acad Sci U S A ; 108(12): 4840-5, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21383185

ABSTRACT

hsa-mir-483 is located within intron 2 of the IGF2 gene. We have previously shown oncogenic features of miR-483-3p through cooperation with IGF2 or by independently targeting the proapoptotic gene BBC3/PUMA. Here we demonstrate that expression of miR-483 can be induced independently of IGF2 by the oncoprotein ß-catenin through an interaction with the basic helix-loop-helix protein upstream stimulatory transcription factor 1. We also show that ß-catenin itself is a target of miR-483-3p, triggering a negative regulatory loop that becomes ineffective in cells harboring an activating mutation of ß-catenin. These results provide insights into the complex regulation of the IGF2/miR-483 locus, revealing players in the ß-catenin pathway.


Subject(s)
MicroRNAs/metabolism , Mutation , beta Catenin/biosynthesis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Genetic Loci/genetics , HEK293 Cells , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Introns/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/genetics
16.
Lancet Oncol ; 14(6): e229-38, 2013 May.
Article in English | MEDLINE | ID: mdl-23639323

ABSTRACT

Protein phosphatase 2A (PP2A), one of the main serine-threonine phosphatases in mammalian cells, maintains cell homoeostasis by counteracting most of the kinase-driven intracellular signalling pathways. Unrestrained activation of oncogenic kinases together with inhibition of tumour suppressors is often required for development of cancer. PP2A has been shown to be genetically altered or functionally inactivated in many solid cancers and leukaemias, and is therefore a tumour suppressor. For example, the phosphatase activity of PP2A is suppressed in chronic myeloid leukaemia and other malignancies characterised by aberrant activity of oncogenic kinases. Preclinical studies show that pharmacological restoration of PP2A tumour-suppressor activity by PP2A-activating drugs (eg, FTY720) effectively antagonises cancer development and progression. Here, we discuss PP2A as a druggable tumour suppressor in view of the possible introduction of PP2A-activating drugs into anticancer therapeutic protocols.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Design , Enzyme Activators/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Protein Phosphatase 2/metabolism , Animals , Enzyme Activation , Humans , Neoplasms/enzymology , Neoplasms/pathology , Signal Transduction/drug effects
17.
J Exp Med ; 204(10): 2397-405, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17875674

ABSTRACT

Monokines (i.e., interleukin [IL]-12, -18, and -15) induce natural killer (NK) cells to produce interferon-gamma (IFN-gamma), which is a critical factor for immune surveillance of cancer and monocyte clearance of infection. We show that SET, which is a potent inhibitor of protein phosphatase type 2A (PP2A) activity, is highly expressed in human CD56bright NK cells, which produce more IFN-gamma than CD56dim NK cells. SET was up-regulated upon monokine stimulation of primary human NK cells. Furthermore, ectopic overexpression of SET significantly enhanced IFN-gamma gene expression in monokine-stimulated NK cells. In contrast, RNAi-mediated suppression of SET expression renders NK cells inefficient in producing high levels of IFN-gamma in response to monokine costimulation. Mechanistically, suppression of PP2A activity by SET is important for IFN-gamma gene expression in NK cells. In fact, treatment of primary human NK cells with the PP2A activator 1,9-dideoxy-forskolin, as well as administration of the drug to C57BL/6 mice, significantly reduced NK-dependent IFN-gamma production in response to monokine treatment. Further, SET knockdown or pharmacologic activation of PP2A diminished extracellular signal-regulated kinase 1/2, p65RelA, signal transducer and activator of transduction 4 (STAT4), and STAT5 activity in monokine-stimulated NK cells, potentially contributing to the reduction in IFN-gamma gene expression. Thus, SET expression is essential for suppressing PP2A phosphatase activity that would otherwise limit NK cell antitumoral and/or antiinflammatory functions by impairing NK cell production of IFN-gamma.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Interferon-gamma/biosynthesis , Killer Cells, Natural/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Enzyme Activation , Gene Expression Regulation , Histone Chaperones , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Monokines/pharmacology , Signal Transduction , Transcription Factors/genetics
19.
Blood ; 117(8): 2378-84, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21156847

ABSTRACT

The ability of natural killer (NK) cells to kill malignant or infected cells depends on the integration of signals from different families of cell surface receptors, including cytokine receptors. How such signals then regulate NK-cell cytotoxicity is incompletely understood. Here we analyzed an endogenous inhibitor of protein phosphatase 2A (PP2A) activity called SET, and its role in regulating human NK-cell cytotoxicity and its mechanism of action in human NK cells. RNAi-mediated suppression of SET down-modulates NK-cell cytotoxicity, whereas ectopic overexpression of SET enhances cytotoxicity. SET knockdown inhibits both mRNA and protein granzyme B expression, as well as perforin expression, whereas SET overexpression enhances granzyme B expression. Treatment of NK cells with the PP2A activator 1,9-dideoxy-forskolin also inhibits both granzyme B expression and cytotoxicity. In addition, pretreatment with the PP2A inhibitor okadaic acid rescues declining granzyme B mRNA levels in SET knockdown cells. Down-modulation of SET expression or activation of PP2A also decreases human NK-cell antibody-dependent cellular cytotoxicity. Finally, the induction of granzyme B gene expression by interleukin-2 and interleukin-15 is inhibited by SET knockdown. These data provide evidence that granzyme B gene expression and therefore human NK-cell cytotoxicity can be regulated by the PP2A-SET interplay.


Subject(s)
Granzymes/genetics , Histone Chaperones/physiology , Killer Cells, Natural/metabolism , Protein Phosphatase 2/physiology , Transcription Factors/physiology , Cytotoxicity, Immunologic , DNA-Binding Proteins , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Granzymes/biosynthesis , Humans , Killer Cells, Natural/immunology , Protein Phosphatase 2/antagonists & inhibitors , RNA, Small Interfering/pharmacology
20.
Blood ; 117(22): 5941-52, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21527515

ABSTRACT

The mechanisms by which sphingosine kinase-1 (SK-1)/sphingosine 1-phosphate (S1P) activation contributes to imatinib resistance in chronic myeloid leukemia (CML) are unknown. We show herein that increased SK-1/S1P enhances Bcr-Abl1 protein stability, through inhibition of its proteasomal degradation in imatinib-resistant K562/IMA-3 and LAMA-4/IMA human CML cells. In fact, Bcr-Abl1 stability was enhanced by ectopic SK-1 expression. Conversely, siRNA-mediated SK-1 knockdown in K562/IMA-3 cells, or its genetic loss in SK-1(-/-) MEFs, significantly reduced Bcr-Abl1 stability. Regulation of Bcr-Abl1 by SK-1/S1P was dependent on S1P receptor 2 (S1P2) signaling, which prevented Bcr-Abl1 dephosphorylation, and degradation via inhibition of PP2A. Molecular or pharmacologic interference with SK-1/S1P2 restored PP2A-dependent Bcr-Abl1 dephosphorylation, and enhanced imatinib- or nilotinib-induced growth inhibition in primary CD34(+) mononuclear cells obtained from chronic phase and blast crisis CML patients, K562/IMA-3 or LAMA4/IMA cells, and 32Dcl3 murine progenitor cells, expressing the wild-type or mutant (Y253H or T315I) Bcr-Abl1 in situ. Accordingly, impaired SK-1/S1P2 signaling enhanced the growth-inhibitory effects of nilotinib against 32D/T315I-Bcr-Abl1-derived mouse allografts. Since SK-1/S1P/S1P2 signaling regulates Bcr-Abl1 stability via modulation of PP2A, inhibition of SK-1/S1P2 axis represents a novel approach to target wild-type- or mutant-Bcr-Abl1 thereby overcoming drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Phosphatase 2/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides , Cell Line, Tumor , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, SCID , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Piperazines/administration & dosage , Protein Phosphatase 2/genetics , Pyrimidines/administration & dosage , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL